Abstract
Ewing sarcoma (ES) is a highly aggressive bone and soft tissue tumor with peak incidence in adolescents and young adults. Despite advances in local control and systemic chemotherapy, metastatic relapse after an initial clinical remission remains a significant clinical problem. In addition, metastasis at the time of presentation or at relapse continues to be the leading cause of death for patients diagnosed with ES. Since the discovery over 20 years ago of the pathognomonic EWS-FLI1 fusion gene, much has been learned about the molecular and cellular biology of ES pathogenesis. In addition, more recent exploitation of advances in stem cell and developmental biology has provided key insights into the cellular origins of ES and the role of epigenetic deregulation in tumor initiation and maintenance. Nevertheless, the mechanisms that drive tumor relapse and metastasis remain largely unknown. These gaps in our knowledge continue to hamper the development of novel therapeutic strategies that will improve outcomes for patients with relapsed and metastatic disease. In this chapter we will review the current status of ES biology research, highlighting areas of investigation that we propose have the greatest potential to yield findings that will translate into clinically significant advances.
Keywords: Ewing sarcoma, metastasis, genetics, epigenetics
I. INTRODUCTION
Ewing sarcoma (ES) is a highly aggressive bone and soft tissue tumor with peak incidence in adolescents and young adults. In the pediatric population it represents the second most common malignant bone tumor, and in adult patients it more commonly presents in soft tissues.1,2 Intensification of systemic chemotherapy and local control measures have improved survival rates for patients with localized tumors to greater than 70%.3 However, event free and overall survival rates for patients with metastatic disease remain dismal at around 20%.4 In addition, for patients who relapse following an initial clinical remission there is little to offer in the way of salvage therapy.5,6 Finally, the acute and long term toxicities associated with current therapeutic regimens are considerable and ES survivors are left with significant health morbidities and shortened life expectancies.7,8 Thus, future improvements in survival and quality of life for patients with ES demand that we better understand the biologic processes that drive tumor relapse and metastasis and that we develop less toxic approaches to ES therapy that will spare normal developing tissues.
In this chapter, we will review the current state of knowledge regarding mechanisms of ES initiation and metastatic progression. We will specifically focus on areas of recent discovery and on identifying areas of investigation that, in our opinion, have the greatest potential to lead to clinically relevant therapeutic advances.
II. TUMOR INITIATION
ES is widely believed to arise from malignant transformation of stem and/or progenitor cells of mesoderm and neural crest lineages.9,10 Specific cells that have been identified as candidate cells of origin include bone marrow-derived mesenchymal stem cells,11,12 neural crest-derived stem cells,13 bone progenitors,14 and most recently, osteochondrogenic progenitors.15 ES tumors express characteristic fusion oncogenes that likely act as the tumor-initiating mutations when expressed in the appropriate cellular and developmental context (see next section and Table 1).16 Given the near universal and ES-specific nature of their expression, detection of EWS-ETS fusions is now routinely used for tumor diagnosis. Studies of the direct and indirect biologic effects of EWS-ETS fusions have provided critical insights into ES pathogenesis and these are discussed below and summarized in Figure 1.
Table 1.
Chromosomal rearrangements found in Ewing sarcoma and Ewing sarcoma-like tumors.
| REARRANGEMENT | FUSION GENE | 
|---|---|
| t(11;22)(q24;q12) | EWSR1-FLI1 | 
| t(21;22)(q22;q12) | EWSR1-ERG | 
| t(7;22)(p22;q12) | EWSR1-ETV1 | 
| t(17;22)(q12;q12) | EWSR1-ETV4 | 
| t(2;22)(q35;q12) | EWSR1-FEV | 
| t(16;21)(p11;q22) | FUS-ERG | 
| t(2;16)(q35;p11) | FUS-FEV | 
| t(20;22)(q13;q12) (NB: can occur in ring chromosome and may be amplified)  | 
EWSR1-NFATC2 | 
| t(6;22)(p21;q12) | EWSR1-POU5F1 | 
| t(4;22)(q31;q12) | 
EWSR1- SMARCA5  | 
| Submicroscopic inv(22) in t(1;22)(p36.1;q12) | EWSR1-ZSG | 
| t(2;22)(q31;q12) | EWSR1-SP3 | 
| t(4;19)(q35;q13) | CIC-DUX4 | 
Figure 1. Model of ES Initiation.
An EWS-ETS fusion gene is created by a chromosomal translocation event during cell division. If the event occurs in a cell type that is tolerant of the fusion oncoprotein, such as a mesenchymal (MSC) or neural crest (NCSC) stem or progenitor cell, in a supportive microenvironment, such as developing bone, tumor initiation can begin. Initiation of malignant transformation downstream of the EWS-ETS fusion gene is dependent on both molecular and cellular changes that, in concert, lead to maintenance of an immature cell state, epigenetic deregulation, and unlimited proliferative capacity. Secondary changes evolve over time that support clonal selection and expansion and ultimately lead to full malignant transformation. These secondary changes likely evolve in response to developmental and growth factor stimuli and occur on a cellular background of epigenetic instability. Latency between the original EWS-ETS fusion event and presentation of ES can be either brief or very prolonged depending on the stochastic nature of secondary changes and their relative potency as pro-oncogenic drivers.
A. Genetics of ES
In ~85-90% of ES cases, a somatic reciprocal t(11;22)(q24;q12) chromosomal translocation is observed that fuses EWSR1 to the FLI1 ETS family gene to generate EWSR1-FLI1 fusion transcripts.17,18 The EWS-FLI1 chimeric oncoprotein is an aberrant transcription factor that deregulates the expression of key genes involved in ES oncogenesis.19,20 Variant EWS-ETS gene fusions are also described, including the t(21;22)(q22;q12) associated EWS-ERG fusion that occurs in 10-15% of cases,21 as well as rarer EWS-ETV1, EWS-ETV4, and EWS-FEV gene fusions.22-25 Rare cases involving FUS, which like EWS, is a member of the FET (FUS, EWS, and TAF15) family of RNA-binding proteins26 have also been described, including FUS-ERG or FUSFEV.27,28 In addition to such FET-ETS gene fusions, cases of so-called “ES-like” tumors are described in which EWS is fused to non-ETS proteins, and others that contain translocations without similarity to EWS-based fusions, such the t(4;19)(q35;q13) generated CIC-DUX4 fusion.16,29-34 How ES-like tumors relate to ES remains contentious.16,35 The current list of ES- and ES-like tumor associated fusions is summarized in Table 1.
Gene fusion detection has emerged as an extremely powerful tool for ES diagnosis. RT-PCR for EWS-FLI1 and EWS-ERG fusions36 theoretically should account for >99% of cases. Fluorescence in situ hybridization (FISH) using an EWS “break-apart” probe or dual color EWS and FLI1 probes is also utilized, but must take into account other EWS-rearranged tumors, such as desmoplastic round cell tumor, myxoid chondrosarcomas, myxoid liposarcoma, or clear cell sarcoma37 Given the above variant fusions, the absence of molecular confirmation of EWS-FLI1 and EWS-ERG fusions may not rule out the diagnosis of ES. Recent next-generation sequencing based approaches may increase the capacity to screen for multiple fusions simultaneously.38,39
Additional chromosomal abnormalities in ES include gains of chromosome 8 in up to 50% of cases,28,40,41 chromosome 12 and 1q gains in 25%,41,42,43 gains in chromosome 20 in 10-20%,44 and 1p36 losses.41 In ~20% of cases, a t(1;16) chromosomal translocation with variable breakpoints occurs and is associated with 1q gains and 16q losses.45-48 Finally, mutations in TP53 and CDKN2A are detected in 10-20% of cases and may be associated with aggressive disease.49-51
B. Biology of EWS-ETS Fusion Proteins
Numerous studies have endeavored to dissect the role of EWS-ETS fusion proteins in ES oncogenesis.52 EWS-ETS chimeric proteins are oncogenic in NIH3T3 fibroblasts 18 and function as aberrant transcription factors binding to ETS consensus sequences of target genes.18,53-55 Among early reported targets of EWS-ETS mediated transcriptional activation are stromelysin 1, cytochrome P-450 F1, cytokeratin 15, manic fringe, E2-C, Id2, PIM3, uridine phosphorylase, and p21WAF1/CIP1.52,56 More recent targets include NKX2.2,19 NROB1,57 GSTM4,58 and STEAP1.59 EWS-FLI1 and other EWS-ETS proteins down-regulate the TGF beta type II receptor (TGFBR2), a putative tumor suppressor gene,60,61 pointing to important roles for EWS-ETS repression of gene subsets. Loss of TGFBR2 expression may provide ES cells with a mechanism to elude programmed cell death. IGFBP-3, an inhibitor of insulin-like growth factor 1 (IGF-1) mediated proliferation and survival signaling, is also a repressed target of EWS-ETS proteins.20 Another mechanism of IGF pathway deregulation in ES appears to be through EWS-FLI1 repression of microRNAs that otherwise negatively regulate the expression of IGF1 and the IGF1 receptor (IGF1R).62 In human mesenchymal stem cells, a potential cell of origin of ES, EWS-FLI1 induces the expression of OCT4, SOX2, and NANOG embryonic stem cell genes12 as well as the polycomb repressor EZH2.63 The significance of these observations in the context of ES biology is under intense investigation and although these discoveries have yielded important insights into disease pathogenesis and new molecular tools for diagnosis, they have not yet significantly impacted ES treatment and outcome.
C. Oncogenic Hubs Downstream of EWS-ETS Fusions
With the advent of genome-wide chromatin immunoprecipitation (ChIP-seq) and RNA interference technologies it has now been established that EWS-FLI1 binds DNA at diverse regions across the genome, including both gene promoter as well as inter- and intra-genic regions,64,65 and that it represses as many genes as it induces.66-68 Thus, EWS-ETS proteins have the potential to modulate gene and protein expression through non-transcriptional mechanisms and, as discussed below, these alternative mechanisms are now recognized to function as critical oncogenic hubs in ES tumorigenesis.
1. Epigenetic Deregulation
Coordinated regulation of gene expression via epigenetic mechanisms is essential for normal development and these regulatory elements are frequently disturbed in both adult and pediatric tumors.69,70 In particular, the actions of polycomb group, histone deacetylase (HDAC) and nucleosome remodeling complexes all contribute to normal regulation of gene expression by altering chromatin structure71,72 and EWS-ETS fusions have been shown to impact on all of these epigenetic hubs.
As mentioned above, the polycomb group protein EZH2 is a direct transcriptional target of EWS-FLI1.63,73 EZH2 represses transcription by methylating histone 3 at lysine residue 27 (H3K27me3) and alterations in H3K27me3 promote oncogenesis.74 In addition, BMI-1, another polycomb group protein, is over-expressed by ES and can be induced by EWS-FLI1, although the mechanism of induction is indirect.13,75,76 BMI-1, in complex with its polycomb partner protein RING1B, ubiquitinates histone 2A, resulting in chromatin compaction and gene silencing.74 The net effect of these polycombdependent modifications is repression of differentiation and maintenance of stemness, two driving forces in the process of malignant transformation.13,63,77
EWS-ETS proteins also alter HDAC-dependent gene regulation. The direct EWS-FLI1 target gene NKX2.2 represses transcription by recruiting transcriptional corepressors and HDACs to gene promoters and many EWS-FLI1-repressed genes are repressed as a consequence of NKX2.2-HDAC-dependent repression.66 Preclinical studies with HDAC inhibitors show anti-tumor effects and therapeutic clinical trials are in development.66,78 In addition, cellular biochemical studies have revealed a key role for HDAC deregulation in the context of the NuRD (nucleosome remodeling and histone deacetylase) complex.79 Repression of the tumor suppressor genes LOX and TGFBR2, among others, is mediated by EWS-FLI1-dependent recruitment of the NuRD complex to gene promoters. Transcriptional repression by NuRD is in part dependent on the histone demethylase, lysine-specific demethylase 1 (LSD1, KDM1A).79,80 LSD1 is over-expressed by ES81 and inhibition of LSD1 in ES by genetic or pharmacologic means results in de-repression of EWS-ETS-suppressed genes and inhibition of tumorigenicity.80 Interestingly, LSD1 inhibition also leads to down-regulation of EWS-FLI1-induced genes demonstrating the complex interplay between chromatin activating and chromatin repressive marks and revealing the central role that EWS-ETS proteins play in disrupting this balance.80 Given its role as a downstream hub of EWS-ETS fusions, and the ongoing efforts by several small and large pharmaceutical companies to develop LSD1 inhibitors, LSD1 is highly promising as a novel target for therapeutic intervention in ES.
Finally, accessibility of transcription factors to DNA is also determined by nucleosome position and structure.72 Studies of chromatin structure in ES cells as well as EWS-FLI1 transduced endothelial cells revealed that the fusion can alter nucleosome occupancy, especially in regions adjacent to its own binding sites.65 This ability of EWS-FLI1 to confer an open chromatin structure indicates that, in addition to altering post-translational modifications of histones, the fusion protein can alter epigenetic regulation of gene expression by interfering with nucleosome remodeling.
2. Aberrant Regulation of RNA Splicing and miRNAs
Early studies of EWS-FLI1 suggested that it may play a role in disrupting RNA splicing.82,83 This initial observation has been supported by new evidence that links both wild-type EWS and EWS-FLI1 to the RNA splicing machinery.84,85 Specifically, wild-type EWS is a key mediator of RNA splicing in response to DNA damage and EWS-FLI1 has been shown to impact expression of splice variants of FAS, Death receptor 4, and VEGF.84,86,87 These studies implicate isoform switching as an indirect mechanism of EWS-ETS-dependent tumorigenesis. Abnormal expression and regulation of numerous microRNAs have also been implicated in disrupted gene expression and ES pathogenesis.62,88-94 In particular, recent studies have revealed a link between EWS-FLI1-dependent disruptions to miRNA processing and maintenance of a cancer stem cell phenotype.95 Specifically, candidate ES cancer stem cells as well as EWS-FLI1-transduced pediatric mesenchymal stem cells display reduced expression of the Dicer complex protein TARBP2, which results in inhibition of miRNA maturation. This fundamental defect in miRNA processing contributes to ES tumorigenicity and provides a potential therapeutic target.95
III. TUMOR METASTASIS
There has been little change in overall outcome in ES for almost twenty years, mainly because we do not yet understand the molecular basis of metastasis in these patients. The presence of metastatic disease is the single-most powerful predictor of outcome in ES and other childhood sarcomas, and the prognosis for patients with metastatic disease remains dismal.96 EWS-ETS gene fusions are present in both localized and widespread disease, and thus they alone cannot account for metastatic behavior.97 There is a compelling need to develop curative treatments for ES patients with metastatic cancer. In particular, micrometastatic disease, representing residual tumor cells in the circulation or various organ sites following removal and treatment of the primary tumor, is a very poorly understood stage in cancer progression. Adjuvant chemotherapy, aimed at preventing such recurrent disease, is conceptually designed to eliminate micrometastases. However, the biology of micrometastatic cells, the purported origin of metastatic disease, remains almost completely unknown. Identifying factors that contribute to micrometastasis and metastatic spread, and that can be targeted therapeutically, have tremendous potential to improve outcome. The multistep process of metastasis has been excellently reviewed elsewhere,98 and this review will instead discuss new concepts in the metastatic cascade that the ES research community needs to be aware of in order to work towards strategies for potentially preventing or targeting the metastatic process in ES (Figure 2).
Figure 2. Biology of ES Progression.
It is likely that all ES cells have the capacity to invade and metastasize as a consequence of the inherent migratory nature of their stem cells of origin. Plasticity in response to cell intrinsic (e.g. metabolic and genotoxic) and cell extrinsic (e.g. hypoxia, nutrient deprivation) stress is a key biologic feature of normal stem cells that is necessary for maintenance of stemness and unlimited proliferative capacity. ES cells, by nature of their cell of origin and epigenetic deregulation, are highly plastic and dynamically respond to stress. Adaptive, reversible responses to stress contribute to metastasis, therapy resistance, and relapse. Epigenetic evolution over time results in the selective outgrowth of clones that have reversibly, or irreversibly, adopted phenotypic changes that are heritably passed on to daughter cells. In this way, metastatic and drug resistant clones emerge in the absence of additional mutation. Finally, irreversible genetic changes that confer a growth advantage, such as loss of tumor suppressor genes or gain of copy number alterations, will be selected for over time and selective pressure for creation and expansion of these clones will be promoted by the DNA damaging effects of chemotherapy.
A. Genomic Studies of Metastatic ES
The advent of next generation sequencing (NGS) methods can potentially shed light into the metastatic cascade in ES through deep sequencing of patient-matched primary and metastatic lesions to identify genetic drivers of metastasis, and this approach has provided novel insights into metastatic disease in adult malignancies.99,100 However, as in other pediatric cancers such as retinoblastoma and neuroblastoma,101-103 the ES genome is proving to be relatively quiet compared to adult malignancies,104,105 and therefore genetic approaches may not delineate the molecular basis of metastatic disease in ES. As will be described in the next section, epigenetic or other mechanisms may instead underlie this process. Moreover, the issue of tumor heterogeneity suggests that analyzing single primary tumor biopsies by NGS may miss metastatic driver mutations,100 i.e. relevant clones may not be present in the tumor section subjected to sequencing in a given NGS experiment. Superimposed on this issue, NGS of pediatric medulloblastoma has revealed that primary and patient-matched metastatic disease represents different genetic compartments; i.e. that advanced medulloblastoma is a bicompartmental disease.106 These studies indicate that metastatic lesions are highly similar to each other across patients, but differ markedly from patient-matched primary tumors, and that only rare clones in the primary tumors are present in the metastases. In fact, so-called post-dispersion events (e.g. mutations or structural variations) may occur after the metastatic clones have migrated from the primary tumor.106 This has major implications for precision medicine, as actionable mutations identified in primary tumors may have no relevance to metastasis, the major cause of death in that patient population. These studies argue for new approaches to better understand metastatic disease in ES. First, clinical practice must strive to include ES biopsies of metastatic lesions for NGS. Second, as a community we must focus more diligently on biology studies that specifically address metastatic processes in ES. Several recent studies in this regard are now discussed.
B. Metastatic Pathways in ES
Surprisingly, few studies have focused on metastatic disease in ES at the mechanistic level, although recent studies have begun to identify candidate proteins and pathways that promote a metastatic ES phenotype. The zinc finger E-box binding homeobox 2 (ZEB2) transcription factor was recently shown to promote metastasis of ES cell lines in vivo and to be more highly expressed by patients with metastatic disease.107 Dickkopf 2 (DKK2) has also been implicated in ES metastasis in vivo, either through activation of its effector matrix metalloproteinase 1 (MMP1), or through upregulation of genes known to be involved in invasion and metastasis such as CXCR4, PTHrP, RUNX2, and TGF β 1.108 CXCR4, a chemokine receptor that is known to bind the CXCL12 chemokine, was also shown to promote migration and invasion of ES cell lines in vitro in a Cdc42 and Rac1 dependent manner.109 CXCR4 expression has previously been associated with ES metastatic disease in patient samples,110 and with ES angiogenesis.111 However, links to metastasis have been disputed by others.112
Analysis of micrometastasis in ES has led to several insights into potential drivers of metastasis. ERBB4 [(HER4) receptor tyrosine kinase, a member of the epidermal growth factor receptor (EGFR) family], activation suppresses anoikis, a hallmark of transformation, in ES cell lines.113 Anoikis, or programmed cell death after detachment from the extracellular matrix,114 is thought to be critical for cancer cells to survive under anchorage independent conditions such as in the circulation or the lymphatics, prior to establishment of overt metastases.115 ES cell lines suppress anoikis and survive as multicellular spheroids in suspension cultures116 in an E-cadherin dependent manner that correlates with increased activation of the PI3 kinase (PI3K)-Akt pathway, upregulation of ERBB4, and enhanced chemoresistance.113 Significantly, ERBB4 is transcriptionally upregulated in cell lines derived from recurrent or metastatic ES, and increased ERBB4 protein expression was identified in metastatic compared to primary patient-matched biopsies and was associated with reduced disease-free survival in clinical cases.117 In addition, ERBB4 overexpression enhanced migration and invasion of ES cells in vitro and increased lung metastases in vivo.117 Upregulation of ERBB4 in ES is dependent on hyperactivation of the PI3K-Akt cascade and on focal adhesion kinase (FAK). Critical for ERBB4-dependent metastasis is the Rac1 GTPase, which is activated through PI3K-Akt and FAK dependent pathways in ES cells in vivo.117 Therefore both DKK2-SDF1α-CXCR4-Rac1 and ERBB4-PI3K-Akt-FAK-Rac1 pathways are potential drivers of metastasis in ES. It will be important to determine whether blocking these pathways pharmacologically, or inhibiting common downstream effectors such as Rac1, can reduce the burden of metastatic disease in ES.
C. Cell Plasticity
Tumor cell heterogeneity that contributes to metastasis can be a result of genetic changes, as discussed above, or non-mutational heterogeneity.118 Heterogeneity that is not driven by differences in DNA sequence is evidenced by inter-cellular differences in gene and/or protein expression and can be mediated by factors ranging from altered epigenetic regulation to altered protein processing.118,119 CXCR4 is heterogeneously expressed in ES and expression is induced under conditions of stress.109 In addition, ERBB4 is preferentially activated under conditions of serum deprivation and anchorage independence.113,117 Thus, these studies together suggest that induction of CXCR4 and ERBB4 may be part of an ES stress response that contributes to tumor metastasis. In breast cancer and other epithelial malignancies, phenotypic transitions between non-invasive and invasive states are mediated in part by epithelial to mesenchymal (EMT) and mesenchymal to epithelial (MET) switches. EMT is induced in response to extracellular signals such as TGF-β and is dependent on epigenetic regulation.120 Given the critical role that epigenetic deregulation plays in ES pathogenesis, it is highly likely that epigenetic switches promote cell state transitions in ES and contribute to ES cell heterogeneity. Determining how metastasis drivers such as CXCR4 and ERBB4 are dynamically induced in ES will support efforts to block these transitions as a novel approach to metastasis inhibition.
The ability of cells to undergo phenotypic switching is a marker of cell plasticity and is a feature of stem and progenitor cells.118 In epithelial tumors there is mounting evidence that cell plasticity is a major determinant of the cancer stem cell phenotype.119,121 Nearly all ES cells robustly express BMI-1, a marker of both normal and cancer stem cells.75,122 Thus, plasticity may be an inherent property of many, if not all, ES cells. As such, ES cells in vivo are also likely to be highly dynamic and responsive to stress. Future studies should focus on developing in vivo and ex vivo models that will permit testing and validation of tumor cell plasticity and its contribution to ES metastasis in real time in contextually relevant microenvironments.
IV. THE WAY FORWARD
Improved outcomes and quality of life for patients remain the ultimate goals of ES research. Unfortunately, the field has reached an impasse largely as a result of a continued inability to improve outcomes for patients with metastatic disease. In this final section we discuss the areas of research that we feel have the greatest potential to yield transformative advances that will translate into benefits for patients.
A. Novel Treatments
Early stage clinical trials continue to test molecularly targeted agents in ES but, to date, results have been largely disappointing.5 Nevertheless, there is mounting preclinical evidence, and some supportive clinical trial data, that targeting the IGF1 pathway and PARP inhibition will be beneficial for at least some patients.5,6 It will be imperative that new agents be tested in combination with other drugs in order to optimize response and minimize emergence of resistant clones.123 Failure to appreciate the absolute nature of emergent resistance to single pathway-targeted therapies will result in failed clinical trials and unwarranted dismissal of promising agents.
Immunotherapy approaches, including PD-1 blockade and chimeric antigen receptor-modified T-cell therapy, are showing tremendous promise in hematologic malignancies as well as some solid tumors.124-126 These findings provide compelling rationale to continue to pursue these avenues in ES, either alone or in combination with epigenetic priming to induce immunogenic antigen expression.127,128 In addition, approaches to directly target EWS-ETS fusions themselves, or their key downstream hubs, should continue as areas of focus.5 With respect to targeting the fusions themselves, small molecule inhibitors of protein-protein interactions, such as YK-4-279129,130 and RNA-interference based technologies131,132 provide proof of concept data that this approach is feasible. With respect to targeting the aforementioned downstream hubs of EWS-ETS action, early preclinical studies with inhibitors of LSD1,80 HDAC,66,133 and DNA methyltransferases,134 suggest that targeting epigenetic deregulation will provide therapeutic benefit. Likewise, targeting of the miRNA machinery has shown promise in preclinical models.95 Continued investigation of hub-targeted therapies should be prioritized given that they are likely to be less susceptible to the development of therapeutic resistance than single pathway-targeted approaches.
B. Novel Models
A critical need is to develop and better utilize models of metastatic ES, such that metastasis inhibition and time to progression studies can be initiated. There is currently a dearth of strategies to assess in vivo metastatic capacity of ES cells. New models for this purpose that should be further developed for these efforts include renal subcapsular implantation117 as well as orthotopic injections into bone.107,135 These models more faithfully recapitulate the metastatic process than traditional subcutaneous or tail vein injection models. In addition, epigenetic instability and evolution of cell lines in culture can result in considerable biologic drift that can mask or even erase key physiologic features of the primary tumor. Moreover, response to therapy can be altered by culture-induced changes. For this reason novel therapies and studies of tumor biology should ideally be performed using early passage cells or patient derived xenografts (PDX).136 Given the rarity of ES and the need for considerable infrastructure to develop PDX models, the ES research community should strive to develop, test, and share these models as a global resource both for studies of tumor biology as well as for testing of novel therapeutic agents and regimens.
Other xenograft models that are worthy of further investigation in ES are the mouse pulmonary metastasis assay (PUMA) and zebrafish metastasis models. In the PUMA assay fluorescently tagged sarcoma cells are injected through the tail vein, and then lungs are cultured ex vivo and assessed for tumor formation.137 In zebrafish, assessment of migration capacity of fluorescence-labeled tumor cells can be determined in real time by in vivo imaging following xenotransplantation of the cells into embryo yolk sacs.138-140 Candidate metastasis genes and pathways that are identified in primary tumors or in vitro assays should be tested in appropriate in vivo models in order to validate their importance to metastatic progression.
Host immune system, cancer associated fibroblasts, blood vessels and other tumor-associated stromal cells contribute to tumorigenicity, tumor progression and drug response.141 Immune deficient animals and other available experimental models of ES largely fail to consider the contribution of these factors and exploration of innovative approaches that utilize so-called humanized mice should be pursued.142 In addition, models that rely on the use of bioengineered 3-dimensional scaffolds have recently been described and provide innovative ex vivo microenvironments in which to study ES tumor cell biology and drug response.143,144 Biologically relevant preclinical models of ES such as these that better recapitulate the cellular and matrix milieu of an established ES tumor should continue to be priorities for research and development. Moreover, preclinical drug testing should optimally be tested in at least one contextually relevant model system in order to improve identification of top promising candidates and expedite successful translation to the clinic.
C. Circulating Tumor Cells
Without systemic chemotherapy localized ES will nearly always recur at distant sites, confirming that micrometastatic disease is essentially universal 145,146 and supporting a need for a better understanding of micrometastatic disease in ES metastasis. A landmark study in pancreatic adenocarcinoma showed that circulating cells with features of EMT can be identified in the circulation in advance of primary tumor detection.147 This suggests that micrometastatic cells may be liberated very early in tumorigenesis, reiterating the need to go beyond the analysis of primary tumors to understand mechanisms of tumor spread. To that end, new technologies in nanomaterials and microfluidics that can isolate circulating tumor cells (CTC),148 tumor exosomes149 and tumor DNA (ctDNA)150,151 should be studied in ES, both in the context of patient-derived peripheral blood samples as well as in animal models of metastasis. Real-time insights into tumor metastasis from its earliest stages will be gained from these studies. In addition, CTC, ctDNA and/or tumor exosomes may prove useful as biomarkers of treatment response and relapse.152
V. CONCLUSION
The persistent failure to improve outcomes for patients with relapsed and metastatic ES demonstrates that much remains to be learned with regards to the biology of tumor progression. In addition, the considerable short- and long-term toxicities that are associated with current chemotherapeutic regimens demand that newer approaches to therapy are developed in order to minimize off-target side effects. Successful integration of novel agents will require their use in the context of rational combination therapies that also consider timing, sequence, and dosing. Ideally, these rational combinations will first be tested in relevant and predictive animal models, thus allowing for rapid prioritization of only the most promising agents and regimens. Finally, there is a critical need for new in vivo and ex vivo models of ES that are more reflective of tumor biology and tumor heterogeneity than current in vitro cell line-based models. With focused efforts to address these gaps in our knowledge the ES research community will be better placed to make discoveries that will favorably impact on patient outcomes.
ACKNOWLEDGEMENTS
The authors would like to acknowledge the many investigators whose work we were unable to cite due to space constraints. The path to improved outcomes for patients with ES has been, and must continue to be, an international effort that depends on the considerable talents and efforts of basic, translational, and clinical investigators across diverse disciplines.
Grant support for this work was provided by 1R01 CA134604 (to ERL) and by funds (to PHS) from the British Columbia Cancer Foundation through generous donations from Team Finn and other riders in the Ride to Conquer Cancer.
ABBREVIATIONS
- BMI-1
 BMI1 proto-oncogene, polycomb ring finger
- CXCR4
 chemokine (C-X-C motif) receptor 4
- EMT
 epithelial to mesenchymal transition
- ERBB4
 v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 4
- ERG
 v-ets avian erythroblastosis virus E26 oncogene homolog
- ES
 Ewing sarcoma
- ETS - E26
 transformation-specific family of transcription factors
- EWSR1
 EWS RNA binding protein 1
- EZH2
 enhancer of zeste 2 polycomb repressive complex 2 subunit
- FET
 family of RNA-binding proteins that includes FUS, EWS, and TAF15
- FLI1
 Fli-1 proto-oncogene
- HDAC
 histone deacetylase
- LSD1 (KDM1A)
 lysine (K)-specific demethylase 1A
- MET
 mesenchymal to epithelial transition
- miRNA
 microRNA
- NKX2.2
 NK2 homeobox 2
- NROB1
 nuclear receptor subfamily 0, group B, member 1
- NURD
 Nucleosome remodeling deacetylase
- TARBP2
 TAR (HIV-1) RNA binding protein 2
- TGFBR2
 Transforming growth factor beta receptor II
REFERENCES
- 1.Karski EE, Matthay KK, Neuhaus JM, Goldsby RE, Dubois SG. Characteristics and outcomes of patients with Ewing sarcoma over 40 years of age at diagnosis. Cancer Epidemiology. 2013;37:29–33. doi: 10.1016/j.canep.2012.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 2.Balamuth NJ, Womer RB. Ewing's sarcoma. Lancet Oncol. 2010;11:184–92. doi: 10.1016/S1470-2045(09)70286-4. [DOI] [PubMed] [Google Scholar]
 - 3.Womer RB, West DC, Krailo MD, Dickman PS, Pawel BR, Grier HE, Marcus K, Sailer S, Healey JH, Dormans JP, Weiss AR. Randomized controlled trial of interval-compressed chemotherapy for the treatment of localized Ewing sarcoma: a report from the Children's Oncology Group. Journal of Clinical Oncology. 2012;30:4148–54. doi: 10.1200/JCO.2011.41.5703. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 4.Grier HE, Krailo MD, Tarbell NJ, Link MP, Fryer CJ, Pritchard DJ, Gebhardt MC, Dickman PS, Perlman EJ, Meyers PA, Donaldson SS, Moore S, Rausen AR, Vietti TJ, Miser JS. Addition of ifosfamide and etoposide to standard chemotherapy for Ewing's sarcoma and primitive neuroectodermal tumor of bone. N Engl J Med. 2003;348:694–701. doi: 10.1056/NEJMoa020890. [DOI] [PubMed] [Google Scholar]
 - 5.Arnaldez FI, Helman LJ. New strategies in ewing sarcoma: lost in translation? Clinical Cancer Research. 2014;20:3050–6. doi: 10.1158/1078-0432.CCR-13-0633. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 6.Kelleher FC, Thomas DM. Molecular pathogenesis and targeted therapeutics in Ewing sarcoma/primitive neuroectodermal tumours. Clinical Sarcoma Research. 2012;2:6. doi: 10.1186/2045-3329-2-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 7.Armstrong GT, Kawashima T, Leisenring W, Stratton K, Stovall M, Hudson MM, Sklar CA, Robison LL, Oeffinger KC. Aging and risk of severe, disabling, life-threatening, and fatal events in the childhood cancer survivor study. Journal of Clinical Oncology. 2014;32:1218–27. doi: 10.1200/JCO.2013.51.1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 8.Ginsberg JP, Goodman P, Leisenring W, Ness KK, Meyers PA, Wolden SL, Smith SM, Stovall M, Hammond S, Robison LL, Oeffinger KC. Long-term survivors of childhood Ewing sarcoma: report from the childhood cancer survivor study. J Natl Cancer Inst. 2010;102:1272–83. doi: 10.1093/jnci/djq278. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 9.Meltzer PS. Is Ewing's sarcoma a stem cell tumor? Cell Stem Cell. 2007;1:13–5. doi: 10.1016/j.stem.2007.05.011. [DOI] [PubMed] [Google Scholar]
 - 10.Riggi N, Suva ML, Stamenkovic I. Ewing's sarcoma origin: from duel to duality. Expert Rev Anticancer Ther. 2009;9:1025–30. doi: 10.1586/era.09.81. [DOI] [PubMed] [Google Scholar]
 - 11.Riggi N, Cironi L, Provero P, Suva ML, Kaloulis K, Garcia-Echeverria C, Hoffmann F, Trumpp A, Stamenkovic I. Development of Ewing's sarcoma from primary bone marrow-derived mesenchymal progenitor cells. Cancer Res. 2005;65:11459–68. doi: 10.1158/0008-5472.CAN-05-1696. [DOI] [PubMed] [Google Scholar]
 - 12.Riggi N, Suva ML, De Vito C, Provero P, Stehle JC, Baumer K, Cironi L, Janiszewska M, Petricevic T, Suva D, Tercier S, Joseph JM, Guillou L, Stamenkovic I. EWS-FLI-1 modulates miRNA145 and SOX2 expression to initiate mesenchymal stem cell reprogramming toward Ewing sarcoma cancer stem cells. Genes Dev. 2010;24:916–32. doi: 10.1101/gad.1899710. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 13.von Levetzow C, Jiang X, Gwye Y, von Levetzow G, Hung L, Cooper A, Hsu JH, Lawlor ER. Modeling initiation of Ewing sarcoma in human neural crest cells. PLoS One. 2011;6:e19305. doi: 10.1371/journal.pone.0019305. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 14.Castillero-Trejo Y, Eliazer S, Xiang L, Richardson JA, Ilaria RL., Jr. Expression of the EWS/FLI-1 oncogene in murine primary bone-derived cells Results in EWS/FLI-1-dependent, ewing sarcoma-like tumors. Cancer Res. 2005;65:8698–705. doi: 10.1158/0008-5472.CAN-05-1704. [DOI] [PubMed] [Google Scholar]
 - 15.Tanaka M, Yamazaki Y, Kanno Y, Igarashi K, Aisaki K, Kanno J, Nakamura T. Ewing's sarcoma precursors are highly enriched in embryonic osteochondrogenic progenitors. The Journal of Clinical Investigation. 2014;124:3061–74. doi: 10.1172/JCI72399. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 16.Sankar S, Lessnick SL. Promiscuous partnerships in Ewing's sarcoma. Cancer Genetics. 2011;204:351–65. doi: 10.1016/j.cancergen.2011.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 17.Delattre O, Zucman J, Plougastel B, Desmaze C, Melot T, Peter M, Kovar H, Joubert I, de Jong P, Rouleau G, Aurias A, Thomas G. Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours. Nature. 1992;359:162–65. doi: 10.1038/359162a0. [DOI] [PubMed] [Google Scholar]
 - 18.May WA, Gishizky ML, Lessnick SL, Lunsford LB, Lewis BC, Delattre O, Zucman J, Thomas G, Denny CT. Ewing sarcoma 11;22 translocation produces a chimeric transcription factor that requires the DNA-binding domain encoded by FLI1 for transformation. Proc Natl Acad Sci USA. 1993;90:5752–56. doi: 10.1073/pnas.90.12.5752. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 19.Smith R, Owen LA, Trem DJ, Wong JS, Whangbo JS, Golub TR, Lessnick SL. Expression profiling of EWS/FLI identifies NKX2.2 as a critical target gene in Ewing's sarcoma. Cancer Cell. 2006;9:405–16. doi: 10.1016/j.ccr.2006.04.004. [DOI] [PubMed] [Google Scholar]
 - 20.Prieur A, Tirode F, Cohen P, Delattre O. EWS/FLI-1 silencing and gene profiling of Ewing cells reveal downstream oncogenic pathways and a crucial role for repression of insulin-like growth factor binding protein 3. Mol Cell Biol. 2004;24:7275–83. doi: 10.1128/MCB.24.16.7275-7283.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 21.Sorensen PH, Lessnick SL, Lopez-Terrada D, Liu XF, Triche TJ, Denny CT. A second Ewing's sarcoma translocation, t(21;22), fuses the EWS gene to another ETS-family transcription factor, ERG. Nat Genet. 1994;6:146–51. doi: 10.1038/ng0294-146. [DOI] [PubMed] [Google Scholar]
 - 22.Jeon IS, Davis JN, Braun BS, Sublett JE, Roussel MF, Denny CT, Shapiro DN. A variant Ewing's sarcoma translocation (7;22) fuses the EWS gene to the ETS gene ETV1. Oncogene. 1995;10:1229–34. [PubMed] [Google Scholar]
 - 23.Kaneko Y, Yoshida K, Handa M, Toyoda Y, Nishihira H, Tanaka Y, Sasaki Y, Ishida S, Higashino F, Fujinaga K. Fusion of an ETS-family gene, EIAF, to EWS by t(17;22)(q12;q12) chromosome translocation in an undifferentiated sarcoma of infancy. Genes Chromosomes Cancer. 1996;15:115–21. doi: 10.1002/(SICI)1098-2264(199602)15:2<115::AID-GCC6>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
 - 24.Peter M, Couturier J, Pacquement H, Michon J, Thomas G, Magdelenat H, Delattre O. A new member of the ETS family fused to EWS in Ewing tumors. Oncogene. 1997;14:1159–64. doi: 10.1038/sj.onc.1200933. [DOI] [PubMed] [Google Scholar]
 - 25.Urano F, Umezawa A, Hong W, Kikuchi H, Hata J. A novel chimera gene between EWS and E1A-F, encoding the adenovirus E1A enhancer-binding protein, in extraosseous Ewing's sarcoma. Biochem Biophys Res Commun. 1996;219:608–12. doi: 10.1006/bbrc.1996.0281. [DOI] [PubMed] [Google Scholar]
 - 26.Tan AY, Manley JL. The TET family of proteins: functions and roles in disease. J Mol Cell Biol. 2009;1:82–92. doi: 10.1093/jmcb/mjp025. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 27.Shing DC, McMullan DJ, Roberts P, Smith K, Chin SF, Nicholson J, Tillman RM, Ramani P, Cullinane C, Coleman N. FUS/ERG gene fusions in Ewing's tumors. Cancer Res. 2003;63:4568–76. [PubMed] [Google Scholar]
 - 28.Ng TL, O'Sullivan MJ, Pallen CJ, Hayes M, Clarkson PW, Winstanley M, Sorensen PH, Nielsen TO, Horsman DE. Ewing sarcoma with novel translocation t(2;16) producing an in-frame fusion of FUS and FEV. J Mol Diagn. 2007;9:459–63. doi: 10.2353/jmoldx.2007.070009. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 29.Kawamura-Saito M, Yamazaki Y, Kaneko K, Kawaguchi N, Kanda H, Mukai H, Gotoh T, Motoi T, Fukayama M, Aburatani H, Takizawa T, Nakamura T. Fusion between CIC and DUX4 up-regulates PEA3 family genes in Ewing-like sarcomas with t(4;19)(q35;q13) translocation. Human Molecular Genetics. 2006;15:2125–37. doi: 10.1093/hmg/ddl136. [DOI] [PubMed] [Google Scholar]
 - 30.Szuhai K, Ijszenga M, de Jong D, Karseladze A, Tanke HJ, Hogendoorn PC. The NFATc2 gene is involved in a novel cloned translocation in a Ewing sarcoma variant that couples its function in immunology to oncology. Clinical Cancer Research. 2009;15:2259–68. doi: 10.1158/1078-0432.CCR-08-2184. [DOI] [PubMed] [Google Scholar]
 - 31.Yamaguchi S, Yamazaki Y, Ishikawa Y, Kawaguchi N, Mukai H, Nakamura T. EWSR1 is fused to POU5F1 in a bone tumor with translocation t(6;22)(p21;q12) Genes Chromosomes Cancer. 2005;43:217–22. doi: 10.1002/gcc.20171. [DOI] [PubMed] [Google Scholar]
 - 32.Sumegi J, Nishio J, Nelson M, Frayer RW, Perry D, Bridge JA. A novel t(4;22)(q31;q12) produces an EWSR1-SMARCA5 fusion in extraskeletal Ewing sarcoma/primitive neuroectodermal tumor. Mod Pathol. 2011;24:333–42. doi: 10.1038/modpathol.2010.201. [DOI] [PubMed] [Google Scholar]
 - 33.Mastrangelo T, Modena P, Tornielli S, Bullrich F, Testi MA, Mezzelani A, Radice P, Azzarelli A, Pilotti S, Croce CM, Pierotti MA, Sozzi G. A novel zinc finger gene is fused to EWS in small round cell tumor. Oncogene. 2000;19:3799–804. doi: 10.1038/sj.onc.1203762. [DOI] [PubMed] [Google Scholar]
 - 34.Wang L, Bhargava R, Zheng T, Wexler L, Collins MH, Roulston D, Ladanyi M. Undifferentiated small round cell sarcomas with rare EWS gene fusions: identification of a novel EWS-SP3 fusion and of additional cases with the EWS-ETV1 and EWS-FEV fusions. J Mol Diagn. 2007;9:498–509. doi: 10.2353/jmoldx.2007.070053. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 35.de Alava E, Lessnick SL, Sorensen PH. Ewing sarcoma. In: Flethcer CM, Bridge JA, Hogendoorn PCW, Mertens F, editors. WHO Classification of Tumours of Sort Tissue and Bone. International Agency for Research on Cancer (IARC); Lyon: 2013. pp. 305–09. [Google Scholar]
 - 36.Delattre O, Zucman J, Melot T, Garau XS, Zucker J-M, Lenoir GM, Ambros PF, Sheer D, Turc-Carel C, Triche TJ, Aurias A, Thomas G. The Ewing family of tumors--A subgroup of small-round-cell tumors defined by specific chimeric transcripts. N Engl J Med. 1994;331:294–99. doi: 10.1056/NEJM199408043310503. [DOI] [PubMed] [Google Scholar]
 - 37.Miettinen M. From morphological to molecular diagnosis of soft tissue tumors. Adv Exp Med Biol. 2006;587:99–113. doi: 10.1007/978-1-4020-5133-3_10. [DOI] [PubMed] [Google Scholar]
 - 38.Qadir MA, Zhan SH, Kwok B, Bruestle J, Drees B, Popescu OE, Sorensen PH. ChildSeq-RNA: A Next-Generation Sequencing-Based Diagnostic Assay to Identify Known Fusion Transcripts in Childhood Sarcomas. J Mol Diagn. 2014;16:361–70. doi: 10.1016/j.jmoldx.2014.01.002. [DOI] [PubMed] [Google Scholar]
 - 39.Sweeney RT, Zhang B, Zhu SX, Varma S, Smith KS, Montgomery SB, van de Rijn M, Zehnder J, West RB. Desktop transcriptome sequencing from archival tissue to identify clinically relevant translocations. The American Journal of Surgical Pathology. 2013;37:796–803. doi: 10.1097/PAS.0b013e31827ad9b2. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 40.Turc-Carel C, Aurias A, Mugneret F, Lizard S, Sidaner I, Volk C, Thiery JP, Olschwang S, Philip I, Berger MP, et al. Chromosomes in Ewing's sarcoma. I. An evaluation of 85 cases of remarkable consistency of t(11;22)(q24;q12) Cancer Genet Cytogenet. 1988;32:229–38. doi: 10.1016/0165-4608(88)90285-3. [DOI] [PubMed] [Google Scholar]
 - 41.Hattinger CM, Rumpler S, Strehl S, Ambros IM, Zoubek A, Potschger U, Gadner H, Ambros PF. Prognostic impact of deletions at 1p36 and numerical aberrations in Ewing tumors. Genes Chromosomes Cancer. 1999;24:243–54. doi: 10.1002/(sici)1098-2264(199903)24:3<243::aid-gcc10>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
 - 42.Armengol G, Tarkkanen M, Virolainen M, Forus A, Valle J, Bohling T, Asko-Seljavaara S, Blomqvist C, Elomaa I, Karaharju E, Kivioja AH, Siimes MA, Tukiainen E, Caballin MR, Myklebost O, Knuutila S. Recurrent gains of 1q, 8 and 12 in the Ewing family of tumours by comparative genomic hybridization. Br J Cancer. 1997;75:1403–9. doi: 10.1038/bjc.1997.242. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 43.Maurici D, Perez-Atayde A, Grier HE, Baldini N, Serra M, Fletcher JA. Frequency and implications of chromosome 8 and 12 gains in Ewing sarcoma. Cancer Genet Cytogenet. 1998;100:106–10. doi: 10.1016/s0165-4608(97)00028-9. [DOI] [PubMed] [Google Scholar]
 - 44.Ozaki T, Paulussen M, Poremba C, Brinkschmidt C, Rerin J, Ahrens S, Hoffmann C, Hillmann A, Wai D, Schaefer KL, Boecker W, Juergens H, Winkelmann W, Dockhorn-Dworniczak B. Genetic imbalances revealed by comparative genomic hybridization in Ewing tumors. Genes Chromosomes Cancer. 2001;32:164–71. doi: 10.1002/gcc.1178. [DOI] [PubMed] [Google Scholar]
 - 45.Douglass EC, Rowe ST, Valentine M, Parham D, Meyer WH, Thompson EI. A second nonrandom translocation, der(16)t(1;16)(q21;q13), in Ewing sarcoma and peripheral neuroectodermal tumor. Cytogenet Cell Genet. 1990;53:87–90. doi: 10.1159/000132901. [DOI] [PubMed] [Google Scholar]
 - 46.Hattinger CM, Rumpler S, Ambros IM, Strehl S, Lion T, Zoubek A, Gadner H, Ambros PF. Demonstration of the translocation der(16)t(1;16)(q12;q11.2) in interphase nuclei of Ewing tumors. Genes Chromosomes Cancer. 1996;17:141–50. doi: 10.1002/(SICI)1098-2264(199611)17:3<141::AID-GCC1>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
 - 47.Mugneret F, Lizard S, Aurias A, Turc-Carel C. Chromosomes in Ewing's sarcoma. II. Nonrandom additional changes, trisomy 8 and der(16)t(1;16) Cancer Genet Cytogenet. 1988;32:239–45. doi: 10.1016/0165-4608(88)90286-5. [DOI] [PubMed] [Google Scholar]
 - 48.Stark B, Mor C, Jeison M, Gobuzov R, Cohen IJ, Goshen Y, Stein J, Fisher S, Ash S, Yaniv I, Zaizov R. Additional chromosome 1q aberrations and der(16)t(1;16), correlation to the phenotypic expression and clinical behavior of the Ewing family of tumors. J Neurooncol. 1997;31:3–8. doi: 10.1023/a:1005731009962. [DOI] [PubMed] [Google Scholar]
 - 49.de Alava E, Antonescu CR, Panizo A, Leung D, Meyers PA, Huvos AG, Pardo-Mindan FJ, Healey JH, Ladanyi M. Prognostic impact of p53 status in Ewing sarcoma. Cancer. 2000;89:783–92. [PubMed] [Google Scholar]
 - 50.Wei G, Antonescu CR, de Alava E, Leung D, Huvos AG, Meyers PA, Healey JH, Ladanyi M. Prognostic impact of INK4A deletion in Ewing sarcoma. Cancer. 2000;89:793–99. doi: 10.1002/1097-0142(20000815)89:4<793::aid-cncr11>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
 - 51.Huang HY, Illei PB, Zhao Z, Mazumdar M, Huvos AG, Healey JH, Wexler LH, Gorlick R, Meyers P, Ladanyi M. Ewing sarcomas with p53 mutation or p16/p14ARF homozygous deletion: a highly lethal subset associated with poor chemoresponse. Journal of Clinical Oncology. 2005;23:548–58. doi: 10.1200/JCO.2005.02.081. [DOI] [PubMed] [Google Scholar]
 - 52.Lessnick SL, Ladanyi M. Molecular pathogenesis of Ewing sarcoma: new therapeutic and transcriptional targets. Annu Rev Pathol. 2012;7:145–59. doi: 10.1146/annurev-pathol-011110-130237. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 53.Ohno T, Rao VN, Reddy SP. EWS/Fli-1 chimeric protein is a transcriptional activator. Cancer Res. 1993;53:5859–63. [PubMed] [Google Scholar]
 - 54.Bailly R-A, Bosselut R, Zucman J, Cornier F, Delattre O, Roussel M, Thomas G, Ghysdael DNA-binding and transcriptional activation properties of the EWS-FLI-1 fusion protein resulting from the t(11;22) translocation in Ewing sarcoma. Mol Cell Biol. 1994;14:3230–41. doi: 10.1128/mcb.14.5.3230. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 55.Lessnick SL, Braun BS, Denny CT, May WA. Multiple domains mediate transformation by the Ewing's sarcoma EWS/FLI-1 fusion gene. Oncogene. 1995;10:423–31. [PubMed] [Google Scholar]
 - 56.Ban J, Siligan C, Kreppel M, Aryee D, Kovar H. EWS-FLI1 in Ewing's sarcoma: real targets and collateral damage. Adv Exp Med Biol. 2006;587:41–52. doi: 10.1007/978-1-4020-5133-3_4. [DOI] [PubMed] [Google Scholar]
 - 57.Boro A, Pretre K, Rechfeld F, Thalhammer V, Oesch S, Wachtel M, Schafer BW, Niggli FK. Small-molecule screen identifies modulators of EWS/FLI1 target gene expression and cell survival in Ewing's sarcoma. International Journal of Cancer Journal International du Cancer. 2012;131:2153–64. doi: 10.1002/ijc.27472. [DOI] [PubMed] [Google Scholar]
 - 58.Luo W, Gangwal K, Sankar S, Boucher KM, Thomas D, Lessnick SL. GSTM4 is a microsatellite-containing EWS/FLI target involved in Ewing's sarcoma oncogenesis and therapeutic resistance. Oncogene. 2009;28:4126–32. doi: 10.1038/onc.2009.262. [DOI] [PubMed] [Google Scholar]
 - 59.Grunewald TG, Diebold I, Esposito I, Plehm S, Hauer K, Thiel U, da Silva-Buttkus P, Neff F, Unland R, Muller-Tidow C, Zobywalski C, Lohrig K, Lewandrowski U, Sickmann A, Prazeres da Costa O, Gorlach A, Cossarizza A, Butt E, Richter GH, Burdach S. STEAP1 is associated with the invasive and oxidative stress phenotype of Ewing tumors. Molecular Cancer Research : MCR. 2012;10:52–65. doi: 10.1158/1541-7786.MCR-11-0524. [DOI] [PubMed] [Google Scholar]
 - 60.Hahm KB, Cho K, Lee C, Im YH, Chang J, Choi SG, Sorensen PH, Thiele CJ, Kim SJ. Repression of the gene encoding the TGF-beta type II receptor is a major target of the EWS-FLI1 oncoprotein [published erratum appears in Nat Genet 1999 Dec;23(4):481] Nat Genet. 1999;23:222–7. doi: 10.1038/13854. [DOI] [PubMed] [Google Scholar]
 - 61.Im YH, Kim HT, Lee C, Poulin D, Welford S, Sorensen PH, Denny CT, Kim SJ. EWS-FLI1, EWS-ERG, and EWS-ETV1 oncoproteins of Ewing tumor family all suppress transcription of transforming growth factor beta type II receptor gene. Cancer Res. 2000;60:1536–40. [PubMed] [Google Scholar]
 - 62.McKinsey EL, Parrish JK, Irwin AE, Niemeyer BF, Kern HB, Birks DK, Jedlicka P. A novel oncogenic mechanism in Ewing sarcoma involving IGF pathway targeting by EWS/Fli1-regulated microRNAs. Oncogene. 2011;30:4910–20. doi: 10.1038/onc.2011.197. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 63.Richter GH, Plehm S, Fasan A, Rossler S, Unland R, Bennani-Baiti IM, Hotfilder M, Lowel D, von Luettichau I, Mossbrugger I, Quintanilla-Martinez L, Kovar H, Staege MS, Muller-Tidow C, Burdach S. EZH2 is a mediator of EWS/FLI1 driven tumor growth and metastasis blocking endothelial and neuro-ectodermal differentiation. Proc Natl Acad Sci U S A. 2009;106:5324–9. doi: 10.1073/pnas.0810759106. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 64.Bilke S, Schwentner R, Yang F, Kauer M, Jug G, Walker RL, Davis S, Zhu YJ, Pineda M, Meltzer PS, Kovar H. Oncogenic ETS fusions deregulate E2F3 target genes in Ewing sarcoma and prostate cancer. Genome Res. 2013;23:1797–809. doi: 10.1101/gr.151340.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 65.Patel M, Simon JM, Iglesia MD, Wu SB, McFadden AW, Lieb JD, Davis IJ. Tumor-specific retargeting of an oncogenic transcription factor chimera results in dysregulation of chromatin and transcription. Genome Research. 2012;22:259–70. doi: 10.1101/gr.125666.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 66.Owen LA, Kowalewski AA, Lessnick SL. EWS/FLI mediates transcriptional repression via NKX2.2 during oncogenic transformation in Ewing's sarcoma. PLoS One. 2008;3:e1965. doi: 10.1371/journal.pone.0001965. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 67.Kauer M, Ban J, Kofler R, Walker B, Davis S, Meltzer P, Kovar H. A molecular function map of Ewing's sarcoma. PLoS One. 2009;4:e5415. doi: 10.1371/journal.pone.0005415. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 68.Stoll G, Surdez D, Tirode F, Laud K, Barillot E, Zinovyev A, Delattre O. Systems biology of Ewing sarcoma: a network model of EWS-FLI1 effect on proliferation and apoptosis. Nucleic Acids Research. 2013;41:8853–71. doi: 10.1093/nar/gkt678. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 69.Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128:683–92. doi: 10.1016/j.cell.2007.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 70.Lawlor ER, Thiele CJ. Epigenetic changes in pediatric solid tumors: promising new targets. Clinical Cancer Research. 2012;18:2768–79. doi: 10.1158/1078-0432.CCR-11-1921. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 71.Laugesen A, Helin K. Chromatin Repressive Complexes in Stem Cells, Development, and Cancer. Cell Stem Cell. 2014;14:735–51. doi: 10.1016/j.stem.2014.05.006. [DOI] [PubMed] [Google Scholar]
 - 72.Voss TC, Hager GL. Dynamic regulation of transcriptional states by chromatin and transcription factors. Nature Reviews Genetics. 2014;15:69–81. doi: 10.1038/nrg3623. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 73.Riggi N, Suva ML, Suva D, Cironi L, Provero P, Tercier S, Joseph JM, Stehle JC, Baumer K, Kindler V, Stamenkovic I. EWS-FLI-1 expression triggers a Ewing's sarcoma initiation program in primary human mesenchymal stem cells. Cancer Res. 2008;68:2176–85. doi: 10.1158/0008-5472.CAN-07-1761. [DOI] [PubMed] [Google Scholar]
 - 74.Bracken AP, Helin K. Polycomb group proteins: navigators of lineage pathways led astray in cancer. Nat Rev Cancer. 2009;9:773–84. doi: 10.1038/nrc2736. [DOI] [PubMed] [Google Scholar]
 - 75.Douglas D, Hsu JH, Hung L, Cooper A, Abdueva D, van Doorninck J, Peng G, Shimada H, Triche TJ, Lawlor ER. BMI-1 promotes Ewing sarcoma tumorigenicity independent of CDKN2A repression. Cancer Res. 2008;68:6507–15. doi: 10.1158/0008-5472.CAN-07-6152. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 76.Zwerner JP, Joo J, Warner KL, Christensen L, Hu-Lieskovan S, Triche TJ, May WA. The EWS/FLI1 oncogenic transcription factor deregulates GLI1. Oncogene. 2008;27:3282–91. doi: 10.1038/sj.onc.1210991. [DOI] [PubMed] [Google Scholar]
 - 77.Burdach S, Plehm S, Unland R, Dirksen U, Borkhardt A, Staege MS, Muller-Tidow C, Richter GH. Epigenetic maintenance of stemness and malignancy in peripheral neuroectodermal tumors by EZH2. Cell Cycle. 2009;8:1991–6. doi: 10.4161/cc.8.13.8929. [DOI] [PubMed] [Google Scholar]
 - 78.Zorzi AP, Bernstein M, Samson Y, Wall DA, Desai S, Nicksy D, Wainman N, Eisenhauer E, Baruchel S. A phase I study of histone deacetylase inhibitor, pracinostat (SB939), in pediatric patients with refractory solid tumors: IND203 a trial of the NCIC IND program/C17 pediatric phase I consortium. Pediatric Blood & Cancer. 2013;60:1868–74. doi: 10.1002/pbc.24694. [DOI] [PubMed] [Google Scholar]
 - 79.Sankar S, Bell R, Stephens B, Zhuo R, Sharma S, Bearss DJ, Lessnick SL. Mechanism and relevance of EWS/FLI-mediated transcriptional repression in Ewing sarcoma. Oncogene. 2013;32:5089–100. doi: 10.1038/onc.2012.525. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 80.Sankar S, Theisen ER, Bearss J, Mulvihill T, Hoffman LM, Sorna V, Beckerle M, Sharma S, Lessnick SL. Reversible LSD1 inhibition interferes with global EWS/ETS transcriptional activity and impedes Ewing sarcoma tumor growth. Clinical Cancer Research. 2014;20:4584–97. doi: 10.1158/1078-0432.CCR-14-0072. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 81.Bennani-Baiti IM, Machado I, Llombart-Bosch A, Kovar H. Lysine-specific demethylase 1 (LSD1/KDM1A/AOF2/BHC110) is expressed and is an epigenetic drug target in chondrosarcoma, Ewing's sarcoma, osteosarcoma, and rhabdomyosarcoma. Human Pathology. 2012;43:1300–7. doi: 10.1016/j.humpath.2011.10.010. [DOI] [PubMed] [Google Scholar]
 - 82.Knoop LL, Baker SJ. EWS/FLI alters 5′-splice site selection. J Biol Chem. 2001;276:22317–22. doi: 10.1074/jbc.M008950200. [DOI] [PubMed] [Google Scholar]
 - 83.Knoop LL, Baker SJ. The splicing factor U1C represses EWS/FLI-mediated transactivation. J Biol Chem. 2000;275:24865–71. doi: 10.1074/jbc.M001661200. [DOI] [PubMed] [Google Scholar]
 - 84.Paronetto MP, Bernardis I, Volpe E, Bechara E, Sebestyen E, Eyras E, Valcarcel J. Regulation of FAS exon definition and apoptosis by the Ewing sarcoma protein. Cell reports. 2014;7:1211–26. doi: 10.1016/j.celrep.2014.03.077. [DOI] [PubMed] [Google Scholar]
 - 85.Paronetto MP, Minana B, Valcarcel J. The Ewing sarcoma protein regulates DNA damage-induced alternative splicing. Molecular Cell. 2011;43:353–68. doi: 10.1016/j.molcel.2011.05.035. [DOI] [PubMed] [Google Scholar]
 - 86.Picarda G, Surget S, Guiho R, Teletchea S, Berreur M, Tirode F, Pellat-Deceunynck C, Heymann D, Trichet V, Redini F. A functional, new short isoform of death receptor 4 in Ewing's sarcoma cell lines may be involved in TRAIL sensitivity/resistance mechanisms. Molecular Cancer Research: MCR. 2012;10:336–46. doi: 10.1158/1541-7786.MCR-11-0390. [DOI] [PubMed] [Google Scholar]
 - 87.Huang G, Zhou Z, Wang H, Kleinerman ES. CAPER-alpha alternative splicing regulates the expression of vascular endothelial growth factor(1)(6)(5) in Ewing sarcoma cells. Cancer. 2012;118:2106–16. doi: 10.1002/cncr.26488. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 88.Iida K, Fukushi J, Matsumoto Y, Oda Y, Takahashi Y, Fujiwara T, Fujiwara-Okada Y, Hatano M, Nabashima A, Kamura S, Iwamoto Y. miR-125b develops chemoresistance in Ewing sarcoma/primitive neuroectodermal tumor. Cancer Cell International. 2013;13:21. doi: 10.1186/1475-2867-13-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 89.Franzetti GA, Laud-Duval K, Bellanger D, Stern MH, Sastre-Garau X, Delattre O. MiR-30a-5p connects EWS-FLI1 and CD99, two major therapeutic targets in Ewing tumor. Oncogene. 2013;32:3915–21. doi: 10.1038/onc.2012.403. [DOI] [PubMed] [Google Scholar]
 - 90.Dylla L, Moore C, Jedlicka P. MicroRNAs in Ewing Sarcoma. Frontiers in Oncology. 2013;3:65. doi: 10.3389/fonc.2013.00065. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 91.Dylla L, Jedlicka P. Growth-promoting role of the miR-106a~363 cluster in Ewing sarcoma. PLoS One. 2013;8:e63032. doi: 10.1371/journal.pone.0063032. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 92.Robin TP, Smith A, McKinsey E, Reaves L, Jedlicka P, Ford HL. EWS/FLI1 regulates EYA3 in Ewing sarcoma via modulation of miRNA-708, resulting in increased cell survival and chemoresistance. Molecular Cancer Research: MCR. 2012;10:1098–108. doi: 10.1158/1541-7786.MCR-12-0086. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 93.De Vito C, Riggi N, Suva ML, Janiszewska M, Horlbeck J, Baumer K, Provero P, Stamenkovic I. Let-7a is a direct EWS-FLI-1 target implicated in Ewing's sarcoma development. PLoS One. 2011;6:e23592. doi: 10.1371/journal.pone.0023592. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 94.Ban J, Jug G, Mestdagh P, Schwentner R, Kauer M, Aryee DN, Schaefer KL, Nakatani F, Scotlandi K, Reiter M, Strunk D, Speleman F, Vandesompele J, Kovar H. Hsa-mir-145 is the top EWS-FLI1-repressed microRNA involved in a positive feedback loop in Ewing's sarcoma. Oncogene. 2011;30:2173–80. doi: 10.1038/onc.2010.581. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 95.De Vito C, Riggi N, Cornaz S, Suva ML, Baumer K, Provero P, Stamenkovic I. A TARBP2-dependent miRNA expression profile underlies cancer stem cell properties and provides candidate therapeutic reagents in Ewing sarcoma. Cancer Cell. 2012;21:807–21. doi: 10.1016/j.ccr.2012.04.023. [DOI] [PubMed] [Google Scholar]
 - 96.Bernstein M, Kovar H, Paulussen M, Randall RL, Schuck A, Teot LA, Juergens H. Ewing's sarcoma family of tumors: current management. Oncologist. 2006;11:503–19. doi: 10.1634/theoncologist.11-5-503. [DOI] [PubMed] [Google Scholar]
 - 97.Ginsberg JP, de Alava E, Ladanyi M, Wexler LH, Kovar H, Paulussen M, Zoubek A, Dockhorn-Dworniczak B, Juergens H, Wunder JS, Andrulis IL, Malik R, Sorensen PH, Womer RB, Barr FG. EWS-FLI1 and EWS-ERG gene fusions are associated with similar clinical phenotypes in Ewing's sarcoma. Journal of Clinical Oncology. 1999;17:1809–14. doi: 10.1200/JCO.1999.17.6.1809. [DOI] [PubMed] [Google Scholar]
 - 98.Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving paradigms. Cell. 2011;147:275–92. doi: 10.1016/j.cell.2011.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 99.Yu M, Ting DT, Stott SL, Wittner BS, Ozsolak F, Paul S, Ciciliano JC, Smas ME, Winokur D, Gilman AJ, Ulman MJ, Xega K, Contino G, Alagesan B, Brannigan BW, Milos PM, Ryan DP, Sequist LV, Bardeesy N, Ramaswamy S, Toner M, Maheswaran S, Haber DA. RNA sequencing of pancreatic circulating tumour cells implicates WNT signalling in metastasis. Nature. 2012;487:510–3. doi: 10.1038/nature11217. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 100.Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, Varela I, Phillimore B, Begum S, McDonald NQ, Butler A, Jones D, Raine K, Latimer C, Santos CR, Nohadani M, Eklund AC, Spencer-Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA, Swanton C. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. The New England Journal of Medicine. 2012;366:883–92. doi: 10.1056/NEJMoa1113205. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 101.Zhang J, Benavente CA, McEvoy J, Flores-Otero J, Ding L, Chen X, Ulyanov A, Wu G, Wilson M, Wang J, Brennan R, Rusch M, Manning AL, Ma J, Easton J, Shurtleff S, Mullighan C, Pounds S, Mukatira S, Gupta P, Neale G, Zhao D, Lu C, Fulton RS, Fulton LL, Hong X, Dooling DJ, Ochoa K, Naeve C, Dyson NJ, Mardis ER, Bahrami A, Ellison D, Wilson RK, Downing JR, Dyer MA. A novel retinoblastoma therapy from genomic and epigenetic analyses. Nature. 2012;481:329–34. doi: 10.1038/nature10733. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 102.Molenaar JJ, Koster J, Zwijnenburg DA, van Sluis P, Valentijn LJ, van der Ploeg I, Hamdi M, van Nes J, Westerman BA, van Arkel J, Ebus ME, Haneveld F, Lakeman A, Schild L, Molenaar P, Stroeken P, van Noesel MM, Ora I, Santo EE, Caron HN, Westerhout EM, Versteeg R. Sequencing of neuroblastoma identifies chromothripsis and defects in neuritogenesis genes. Nature. 2012;483:589–93. doi: 10.1038/nature10910. [DOI] [PubMed] [Google Scholar]
 - 103.Pugh TJ, Morozova O, Attiyeh EF, Asgharzadeh S, Wei JS, Auclair D, Carter SL, Cibulskis K, Hanna M, Kiezun A, Kim J, Lawrence MS, Lichenstein L, McKenna A, Pedamallu CS, Ramos AH, Shefler E, Sivachenko A, Sougnez C, Stewart C, Ally A, Birol I, Chiu R, Corbett RD, Hirst M, Jackman SD, Kamoh B, Khodabakshi AH, Krzywinski M, Lo A, Moore RA, Mungall KL, Qian J, Tam A, Thiessen N, Zhao Y, Cole KA, Diamond M, Diskin SJ, Mosse YP, Wood AC, Ji L, Sposto R, Badgett T, London WB, Moyer Y, Gastier-Foster JM, Smith MA, Guidry Auvil JM, Gerhard DS, Hogarty MD, Jones SJ, Lander ES, Gabriel SB, Getz G, Seeger RC, Khan J, Marra MA, Meyerson M, Maris JM. The genetic landscape of high-risk neuroblastoma. Nature Genetics. 2013;45:279–84. doi: 10.1038/ng.2529. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 104.Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, Carter SL, Stewart C, Mermel CH, Roberts SA, Kiezun A, Hammerman PS, McKenna A, Drier Y, Zou L, Ramos AH, Pugh TJ, Stransky N, Helman E, Kim J, Sougnez C, Ambrogio L, Nickerson E, Shefler E, Cortes ML, Auclair D, Saksena G, Voet D, Noble M, DiCara D, Lin P, Lichtenstein L, Heiman DI, Fennell T, Imielinski M, Hernandez B, Hodis E, Baca S, Dulak AM, Lohr J, Landau DA, Wu CJ, Melendez-Zajgla J, Hidalgo-Miranda A, Koren A, McCarroll SA, Mora J, Lee RS, Crompton B, Onofrio R, Parkin M, Winckler W, Ardlie K, Gabriel SB, Roberts CW, Biegel JA, Stegmaier K, Bass AJ, Garraway LA, Meyerson M, Golub TR, Gordenin DA, Sunyaev S, Lander ES, Getz G. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499:214–8. doi: 10.1038/nature12213. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 105.Brohl AS, Solomon DA, Chang W, Wang J, Song Y, Sindiri S, Patidar R, Hurd L, Chen L, Shern JF, Liao H, Wen X, Gerard J, Kim JS, Lopez Guerrero JA, Machado I, Wai DH, Picci P, Triche T, Horvai AE, Miettinen M, Wei JS, Catchpool D, Llombart-Bosch A, Waldman T, Khan J. The Genomic Landscape of the Ewing Sarcoma Family of Tumors Reveals Recurrent STAG2 Mutation. PLoS Genetics. 2014;10:e1004475. doi: 10.1371/journal.pgen.1004475. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 106.Wu X, Northcott PA, Dubuc A, Dupuy AJ, Shih DJ, Witt H, Croul S, Bouffet E, Fults DW, Eberhart CG, Garzia L, Van Meter T, Zagzag D, Jabado N, Schwartzentruber J, Majewski J, Scheetz TE, Pfister SM, Korshunov A, Li XN, Scherer SW, Cho YJ, Akagi K, MacDonald TJ, Koster J, McCabe MG, Sarver AL, Collins VP, Weiss WA, Largaespada DA, Collier LS, Taylor MD. Clonal selection drives genetic divergence of metastatic medulloblastoma. Nature. 2012;482:529–33. doi: 10.1038/nature10825. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 107.Wiles ET, Bell R, Thomas D, Beckerle M, Lessnick SL. ZEB2 Represses the Epithelial Phenotype, Facilitates Metastasis in Ewing Sarcoma. Genes & Cancer. 2013;4:486–500. doi: 10.1177/1947601913506115. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 108.Hauer K, Calzada-Wack J, Steiger K, Grunewald TG, Baumhoer D, Plehm S, Buch T, Prazeres da Costa O, Esposito I, Burdach S, Richter GH. DKK2 mediates osteolysis, invasiveness, and metastatic spread in Ewing sarcoma. Cancer Res. 2013;73:967–77. doi: 10.1158/0008-5472.CAN-12-1492. [DOI] [PubMed] [Google Scholar]
 - 109.Krook MA, Nicholls LA, Scannell CA, Chugh R, Thomas DG, Lawlor ER. Stress-induced CXCR4 Promotes Migration and Invasion of Ewing Sarcoma. Molecular Cancer Research: MCR. 2014;12:953–64. doi: 10.1158/1541-7786.MCR-13-0668. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 110.Bennani-Baiti IM, Cooper A, Lawlor ER, Kauer M, Ban J, Aryee DN, Kovar H. Intercohort gene expression co-analysis reveals chemokine receptors as prognostic indicators in Ewing's sarcoma. Clinical Cancer Research. 2010;16:3769–78. doi: 10.1158/1078-0432.CCR-10-0558. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 111.Hamdan R, Zhou Z, Kleinerman ES. Blocking SDF-1alpha/CXCR4 downregulates PDGF-B and inhibits bone marrow-derived pericyte differentiation and tumor vascular expansion in Ewing tumors. Molecular Cancer Therapeutics. 2014;13:483–91. doi: 10.1158/1535-7163.MCT-13-0447. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 112.Berghuis D, Schilham MW, Santos SJ, Savola S, Knowles HJ, Dirksen U, Schaefer KL, Vakkila J, Hogendoorn PC, Lankester AC. The CXCR4-CXCL12 axis in Ewing sarcoma: promotion of tumor growth rather than metastatic disease. Clinical Sarcoma Research. 2012;2:24. doi: 10.1186/2045-3329-2-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 113.Kang HG, Jenabi JM, Zhang J, Keshelava N, Shimada H, May WA, Ng T, Reynolds CP, Triche TJ, Sorensen PH. E-cadherin cell-cell adhesion in ewing tumor cells mediates suppression of anoikis through activation of the ErbB4 tyrosine kinase. Cancer Res. 2007;67:3094–105. doi: 10.1158/0008-5472.CAN-06-3259. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 114.Frisch SM, Screaton RA. Anoikis mechanisms. Curr Opin Cell Biol. 2001;13:555–62. doi: 10.1016/s0955-0674(00)00251-9. [DOI] [PubMed] [Google Scholar]
 - 115.Simpson CD, Anyiwe K, Schimmer AD. Anoikis resistance and tumor metastasis. Cancer Letters. 2008;272:177–85. doi: 10.1016/j.canlet.2008.05.029. [DOI] [PubMed] [Google Scholar]
 - 116.Lawlor ER, Scheel C, Irving J, Sorensen PH. Anchorage-independent multicellular spheroids as an in vitro model of growth signaling in Ewing tumors. Oncogene. 2002;21:307–18. doi: 10.1038/sj.onc.1205053. [DOI] [PubMed] [Google Scholar]
 - 117.Mendoza-Naranjo A, El-Naggar A, Wai DH, Mistry P, Lazic N, Ayala FR, da Cunha IW, Rodriguez-Viciana P, Cheng H, Tavares Guerreiro Fregnani JH, Reynolds P, Arceci RJ, Nicholson A, Triche TJ, Soares FA, Flanagan AM, Wang YZ, Strauss SJ, Sorensen PH. ERBB4 confers metastatic capacity in Ewing sarcoma. EMBO Molecular Medicine. 2013;5:1019–34. doi: 10.1002/emmm.201202343. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 118.Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell plasticity. Nature. 2013;501:328–37. doi: 10.1038/nature12624. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 119.Easwaran H, Tsai HC, Baylin SB. Cancer Epigenetics: Tumor Heterogeneity, Plasticity of Stem-like States, and Drug Resistance. Molecular Cell. 2014;54:716–27. doi: 10.1016/j.molcel.2014.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 120.Chaffer CL, Marjanovic ND, Lee T, Bell G, Kleer CG, Reinhardt F, D'Alessio AC, Young RA, Weinberg RA. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell. 2013;154:61–74. doi: 10.1016/j.cell.2013.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 121.Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 2010;29:4741–51. doi: 10.1038/onc.2010.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 122.Cooper A, van Doorninck J, Ji L, Russell D, Ladanyi M, Shimada H, Krailo M, Womer RB, Hsu JH, Thomas D, Triche TJ, Sposto R, Lawlor ER. Ewing tumors that do not overexpress BMI-1 are a distinct molecular subclass with variant biology: a report from the Children's Oncology Group. Clinical Cancer Research. 2011;17:56–66. doi: 10.1158/1078-0432.CCR-10-1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 123.Lackner MR, Wilson TR, Settleman J. Mechanisms of acquired resistance to targeted cancer therapies. Future Oncology. 2012;8:999–1014. doi: 10.2217/fon.12.86. [DOI] [PubMed] [Google Scholar]
 - 124.Leuci V, Mesiano G, Gammaitoni L, Aglietta M, Sangiolo D. Genetically redirected T lymphocytes for adoptive immunotherapy of solid tumors. Current Gene Therapy. 2014;14:52–62. doi: 10.2174/1566523213666131223130353. [DOI] [PubMed] [Google Scholar]
 - 125.Barrett DM, Singh N, Porter DL, Grupp SA, June CH. Chimeric antigen receptor therapy for cancer. Annual Review of Medicine. 2014;65:333–47. doi: 10.1146/annurev-med-060512-150254. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 126.Dolan DE, Gupta S. PD-1 Pathway Inhibitors: Changing the Landscape of Cancer Immunotherapy. Cancer Control: Journal of the Moffitt Cancer Center. 2014;21:231–7. doi: 10.1177/107327481402100308. [DOI] [PubMed] [Google Scholar]
 - 127.Li H, Chiappinelli KB, Guzzetta AA, Easwaran H, Yen RW, Vatapalli R, Topper MJ, Luo J, Connolly RM, Azad NS, Stearns V, Pardoll DM, Davidson N, Jones PA, Slamon DJ, Baylin SB, Zahnow CA, Ahuja N. Immune regulation by low doses of the DNA methyltransferase inhibitor 5-azacitidine in common human epithelial cancers. Oncotarget. 2014;5:587–98. doi: 10.18632/oncotarget.1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 128.Krishnadas DK, Bao L, Bai F, Chencheri SC, Lucas K. Decitabine facilitates immune recognition of sarcoma cells by upregulating CT antigens, MHC molecules, and ICAM-1. Tumour biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine. 2014;35:5753–62. doi: 10.1007/s13277-014-1764-9. [DOI] [PubMed] [Google Scholar]
 - 129.Hong SH, Youbi SE, Hong SP, Kallakury B, Monroe P, Erkizan HV, Barber-Rotenberg JS, Houghton P, Uren A, Toretsky JA. Pharmacokinetic modeling optimizes inhibition of the 'undruggable' EWS-FLI1 transcription factor in Ewing Sarcoma. Oncotarget. 2014;5:338–50. doi: 10.18632/oncotarget.1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 130.Erkizan HV, Kong Y, Merchant M, Schlottmann S, Barber-Rotenberg JS, Yuan L, Abaan OD, Chou TH, Dakshanamurthy S, Brown ML, Uren A, Toretsky JA. A small molecule blocking oncogenic protein EWS-FLI1 interaction with RNA helicase A inhibits growth of Ewing's sarcoma. Nature Medicine. 2009;15:750–6. doi: 10.1038/nm.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 131.Alhaddad A, Durieu C, Dantelle G, Le Cam E, Malvy C, Treussart F, Bertrand JR. Influence of the internalization pathway on the efficacy of siRNA delivery by cationic fluorescent nanodiamonds in the Ewing sarcoma cell model. PLoS One. 2012;7:e52207. doi: 10.1371/journal.pone.0052207. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 132.Takigami I, Ohno T, Kitade Y, Hara A, Nagano A, Kawai G, Saitou M, Matsuhashi A, Yamada K, Shimizu K. Synthetic siRNA targeting the breakpoint of EWS/Fli-1 inhibits growth of Ewing sarcoma xenografts in a mouse model. International Journal of Cancer Journal International du Cancer. 2011;128:216–26. doi: 10.1002/ijc.25564. [DOI] [PubMed] [Google Scholar]
 - 133.Sonnemann J, Dreyer L, Hartwig M, Palani CD, Hong le TT, Klier U, Broker B, Volker U, Beck JF. Histone deacetylase inhibitors induce cell death and enhance the apoptosis-inducing activity of TRAIL in Ewing's sarcoma cells. Journal of Cancer Research and Clinical Oncology. 2007;133:847–58. doi: 10.1007/s00432-007-0227-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 134.Hurtubise A, Bernstein ML, Momparler RL. Preclinical evaluation of the antineoplastic action of 5-aza-2′-deoxycytidine and different histone deacetylase inhibitors on human Ewing's sarcoma cells. Cancer Cell International. 2008;8:16. doi: 10.1186/1475-2867-8-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 135.Guan H, Zhou Z, Gallick GE, Jia SF, Morales J, Sood AK, Corey SJ, Kleinerman ES. Targeting Lyn inhibits tumor growth and metastasis in Ewing's sarcoma. Molecular Cancer Therapeutics. 2008;7:1807–16. doi: 10.1158/1535-7163.MCT-08-0058. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 136.Stebbing J, Paz K, Schwartz GK, Wexler LH, Maki R, Pollock RE, Morris R, Cohen R, Shankar A, Blackman G, Harding V, Vasquez D, Krell J, Ciznadija D, Katz A, Sidransky D. Patient-derived xenografts for individualized care in advanced sarcoma. Cancer. 2014;120:2006–15. doi: 10.1002/cncr.28696. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 137.Mendoza A, Hong SH, Osborne T, Khan MA, Campbell K, Briggs J, Eleswarapu A, Buquo L, Ren L, Hewitt SM, Dakir el H, Garfield S, Walker R, Merlino G, Green JE, Hunter KW, Wakefield LM, Khanna C. Modeling metastasis biology and therapy in real time in the mouse lung. The Journal of Clinical Investigation. 2010;120:2979–88. doi: 10.1172/JCI40252. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 138.Konantz M, Balci TB, Hartwig UF, Dellaire G, Andre MC, Berman JN, Lengerke C. Zebrafish xenografts as a tool for in vivo studies on human cancer. Annals of the New York Academy of Sciences. 2012;1266:124–37. doi: 10.1111/j.1749-6632.2012.06575.x. [DOI] [PubMed] [Google Scholar]
 - 139.Daugaard M, Nitsch R, Razaghi B, McDonald L, Jarrar A, Torrino S, Castillo-Lluva S, Rotblat B, Li L, Malliri A, Lemichez E, Mettouchi A, Berman JN, Penninger JM, Sorensen PH. Hace1 controls ROS generation of vertebrate Rac1-dependent NADPH oxidase complexes. Nature Communications. 2013;4:2180. doi: 10.1038/ncomms3180. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 140.Veinotte CJ, Dellaire G, Berman JN. Hooking the big one: the potential of zebrafish xenotransplantation to reform cancer drug screening in the genomic era. Disease Models & Mechanisms. 2014;7:745–54. doi: 10.1242/dmm.015784. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 141.Holzel M, Bovier A, Tuting T. Plasticity of tumour and immune cells: a source of heterogeneity and a cause for therapy resistance? Nat Rev Cancer. 2013;13:365–76. doi: 10.1038/nrc3498. [DOI] [PubMed] [Google Scholar]
 - 142.Thibaudeau L, Taubenberger AV, Holzapfel BM, Quent VM, Fuehrmann T, Hesami P, Brown TD, Dalton PD, Power CA, Hollier BG, Hutmacher DW. A tissue-engineered humanized xenograft model of human breast cancer metastasis to bone. Disease models & Mechanisms. 2014;7:299–309. doi: 10.1242/dmm.014076. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 143.Fong EL, Lamhamedi-Cherradi SE, Burdett E, Ramamoorthy V, Lazar AJ, Kasper FK, Farach-Carson MC, Vishwamitra D, Demicco EG, Menegaz BA, Amin HM, Mikos AG, Ludwig JA. Modeling Ewing sarcoma tumors in vitro with 3D scaffolds. Proc Natl Acad Sci U S A. 2013;110:6500–5. doi: 10.1073/pnas.1221403110. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 144.Villasante A, Marturano-Kruik A, Vunjak-Novakovic G. Bioengineered human tumor within a bone niche. Biomaterials. 2014;35:5785–94. doi: 10.1016/j.biomaterials.2014.03.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 145.Wang CC, Schulz MD. Ewing's sarcoma; a study of fifty cases treated at the Massachusetts General Hospital, 1930-1952 inclusive. N Engl J Med. 1953;248:571–6. doi: 10.1056/NEJM195304022481401. [DOI] [PubMed] [Google Scholar]
 - 146.Dahlin DC, Coventry MB, Scanlon PW. Ewing's sarcoma. A critical analysis of 165 cases. The Journal of Bone and Joint Surgery American. 1961;43-A:185–92. [PubMed] [Google Scholar]
 - 147.Rhim AD, Mirek ET, Aiello NM, Maitra A, Bailey JM, McAllister F, Reichert M, Beatty GL, Rustgi AK, Vonderheide RH, Leach SD, Stanger BZ. EMT and dissemination precede pancreatic tumor formation. Cell. 2012;148:349–61. doi: 10.1016/j.cell.2011.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 148.Satelli A, Mitra A, Cutrera JJ, Devarie M, Xia X, Ingram DR, Dibra D, Somaiah N, Torres KE, Ravi V, Ludwig JA, Kleinerman ES, Li S. Universal marker and detection tool for human sarcoma circulating tumor cells. Cancer Res. 2014;74:1645–50. doi: 10.1158/0008-5472.CAN-13-1739. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 149.Miller IV, Raposo G, Welsch U, Prazeres da Costa O, Thiel U, Lebar M, Maurer M, Bender HU, von Luettichau I, Richter GH, Burdach S, Grunewald TG. First identification of Ewing's sarcoma-derived extracellular vesicles and exploration of their biological and potential diagnostic implications. Biology of the Cell / under the auspices of the European Cell Biology Organization. 2013;105:289–303. doi: 10.1111/boc.201200086. [DOI] [PubMed] [Google Scholar]
 - 150.Yu M, Wan YF, Zou QH. Cell-free circulating mitochondrial DNA in the serum: a potential non-invasive biomarker for Ewing's sarcoma. Archives of Medical Research. 2012;43:389–94. doi: 10.1016/j.arcmed.2012.06.007. [DOI] [PubMed] [Google Scholar]
 - 151.Esposito A, Bardelli A, Criscitiello C, Colombo N, Gelao L, Fumagalli L, Minchella I, Locatelli M, Goldhirsch A, Curigliano G. Monitoring tumor-derived cell-free DNA in patients with solid tumors: clinical perspectives and research opportunities. Cancer Treatment Reviews. 2014;40:648–55. doi: 10.1016/j.ctrv.2013.10.003. [DOI] [PubMed] [Google Scholar]
 - 152.Khoja L, Lorigan P, Dive C, Keilholz U, Fusi A. Circulating tumor cells as tumor biomarkers in melanoma: detection methods and clinical relevance. Annals of Oncology. 2015;26:33–9. doi: 10.1093/annonc/mdu207. [DOI] [PubMed] [Google Scholar]
 


