Published OnlineFirst April 22, 2014; DOI: 10.1158/1078-0432.CCR-13-0633

Clinical CCR New Strategies Research

New Strategies in Ewing Sarcoma: Lost in Translation?

Fernanda I. Arnaldez and Lee J. Helman

Abstract Ewing sarcoma is the second most common pediatric malignant bone tumor. Aggressive multimodality therapy has led to an improvement in outcomes, particularly in patients with localized disease. However, therapy-related toxicities are not trivial, and the prognosis for patients with relapsed and/or metastatic disease continues to be poor. In this article, we outline some of the promising therapies that have the potential to change the Ewing sarcoma therapeutic paradigm in the not-too-distant future: insulin-like growth factor receptor inhibitors, targeting of the fusion protein, epigenetic manipulation, PARP inhibitors, and immunotherapy. Clin Cancer Res; 20(12); 1–7. Ó2014 AACR.

Background other fusions have been described (10, 11). This fusion gives Ewing sarcoma is the second most common pediatric origin to a chimeric transcription factor that is responsible malignant bone tumor, and it can also present as a soft- for the Ewing sarcoma oncogenic program. This aberrant tissue malignancy. Approximately 225 new cases are diag- factor modifies the transcription machinery, activating or nosed each year in the United States in patients between the quite often repressing transcription of target genes (12, 13). ages of 1 and 20 years (1). The presence of macrometastatic Although the cell of origin has been much debated, growing disease continues to be the single most significant predictor evidence suggests that Ewing sarcoma could possibly orig- of poor clinical outcome (2): Patients with localized disease inate in mesenchymal stem cells (14). treated with multimodality therapy can achieve a 5-year Despite a growing body of knowledge about Ewing event-free survival rate of 70% (3, 4), whereas the 5-year sarcoma biology, the successful application of basic discov- overall survival rate of patients who present with overt bone eries to the clinic has remained elusive. Preclinical results or bone marrow metastatic disease at diagnosis is less than have not always been predictive of clinical trial outcomes, 20% (1, 5, 6). Traditional therapeutic approaches include highlighting the need for better models with the capability local control of the primary lesion that involves surgery to identify targetable drivers of disease. Improved preclin- and/or radiation therapy, and treatment of disseminated ical models and application of innovative clinical trial disease with multiagent cytotoxic . designs are needed, including the incorporation of combi- These approaches have led to significant improvements natorial therapy in early-phase therapeutic development. in outcomes over the past decades, particularly in patients However, several recent contributions hold promise for the with localized disease (7). However, novel therapeutic future and are discussed below (Table 1). approaches are clearly needed, not only to increase survival in patients with relapsed or metastatic disease (2), but also On the Horizon to continue to improve survival of patients with localized Targeting the EWS–Fli1 fusion protein disease as well as to decrease the acute and chronic toxicities Efforts to suppress the oncogenicity of the fusion protein associated with current cytotoxic drugs. can be described in two fronts: inhibition of the transcrip- The molecular hallmark of Ewing sarcoma is the trans- tion factor activity and inhibition of selected downstream location between EWS, an FET family protein, and an ETS target genes (Fig. 1). The discovery of the EWSR1–Fli1 (EF) transcription factor (8). The FET family includes nuclear fusion protein was reported in 1992 (8). This fusion protein proteins such as FUS, EWS, and TAF15, which are involved generated by the Ewing-specific t(11;22) translocation func- in abnormal rearrangements with transcription factors (9). tions as an aberrant transcription factor that leads to an In 85% of cases, the t(11;22)(q24;q12) translocation altered transcriptional profile of both upregulated and between the EWSR1 and FLI1 is detected. However, many downregulated transcripts (12, 13). Preclinical models sup- port the potential of the EF fusion protein as a therapeutic target in Ewing sarcoma. Synthetic RNAi targeting of the in vivo in vitro Authors' Affiliation: Pediatric Oncology Branch, National Cancer Institute, fusion led to inhibition of tumor growth and . NIH, Bethesda, Maryland In these models, RNAi knockdown was associated with Corresponding Author: Fernanda I. Arnaldez, National Cancer Institute, decreased expression of the EF fusion protein and down- 10 Center Drive, Bldg 10CRC Room 13816, Bethesda, MD 20892. Phone: regulation of EF transcriptional targets such as c-Myc (15). 301-451-7014; Fax: 301-451-7010; E-mail: [email protected] Inhibition or knockdown of EWS–Fli1 in vitro leads to doi: 10.1158/1078-0432.CCR-13-0633 decreased cell viability in vitro (16). Concurrent adminis- Ó2014 American Association for Cancer Research. tration of rapamycin and antisense oligonucleotides

www.aacrjournals.org OF1

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst April 22, 2014; DOI: 10.1158/1078-0432.CCR-13-0633

Arnaldez and Helman

Table 1. Novel therapeutic approaches in Ewing sarcoma

Outcome for phase Strategy Agent Development II studies Refs. Inhibition of EF RNAi Preclinical (14, 15, 17, 19) YK-4-279 Inhibition of EF transcriptional Cytarabine Clinical (phase II) Negative (22) signature Trabectedine Clinical (phase II) Negative (23, 24) Mithramycin Clinical (phase I/II) Ongoing (26) Midostaurin Preclinical (25) Inhibition of tumor growth signaling Insulin growth factor receptor I blockade Clinical (phase II) Positive (34) pathways (antibodies, kinase inhibitors) Disruption of DNA repair PARP inhibitors Clinical (phase II) Negative (45) Epigenetic deregulation HDAC inhibitors (vorinostat) Clinical (phase I) (53, 56) Immune-based therapies Lexatumumab Clinical (phase I) (60) NK cell–based therapies Clinical (phase I) (65, 66) CAR Preclinical (67, 68)

Abbreviations: CAR, chimeric antigen receptor; HDAC, histone deacetylase.

showed delayed tumor growth in murine xenografts (17). tional activity (22). Unfortunately, cytarabine did not show Although these results are encouraging, in vivo delivery of benefit in a phase II clinical trial and was associated with RNAi has proved to be challenging. Using a synthetic significant hematologic toxicity (23). In a similar fashion, polymeric nanoparticle as a carrier, an RNAi directed although preclinical work showed that trabectedin could toward the EWS–Fli1 fusion is being studied in murine interfere with the transcriptional activity of EWS–Fli1 (24), xenografts. The strategy used to target tumor cells is to only 1 patient with Ewing sarcoma achieved stable disease couple the siRNA-carrying nanoparticles with a monoclo- (SD) in a recent phase II trial (25). nal antibody against CD99, an antigen widely expressed in More recently, 1,280 compounds were functionally the surface of Ewing sarcoma cells. Efficacy measures of this screened for suppression of EWS–Fli1 activity. Midostaurin, creative approach will be of interest in the near future (18). a multikinase inhibitor, was shown to modulate the expres- Thus, it has become clear that the EF fusion transcription sion of EWS–Fli1 target genes, and it was associated with factor is necessary for tumor growth, and targeting the decreased tumor growth in xenografts (26). Although mid- mutant transcription factor itself or critical downstream ostaurin is undergoing clinical evaluation for the treatment targets of this protein are attractive therapeutic strategies. of hematologic malignancies, no clinical data in solid However, direct inhibition of transcription factors has tumors are available at this time. Finally, a library of proved to be difficult. The fusion protein has been shown 50,000 compounds was functionally screened using a to interact with RNA helicase A (RHA) using phage display reporter of EWS transcriptional activity (27), and mithra- and chromatin immunoprecipitation techniques (19). Fol- mycin, an antineoplastic antibiotic, was identified as the low-up studies showed that RHA stimulated the activity leading hit. Interestingly, reports of Ewing sarcoma of promoters regulated by EWS–Fli1. Using Plasmon reso- responses to this drug had been reported decades ago nance screening, YK-4-279, an inhibitor of the EWS–Fli1– (28). Mithramycin treatment led to decreased tumor growth RHA interaction has been identified. This compound was in xenograft models, as well as inhibition of the EWS–Fli1 associated with decreased tumor growth in orthotopic signature. A phase I/II clinical trial evaluating the activity of xenografts, but has not progressed to clinical development mithramycin in patients with Ewing sarcoma is ongoing. yet (20, 21). The transcriptional signature of the EF fusion has been Insulin-like growth factor receptor blockade described (12), and as noted above, another way to target The relevance of insulin-like growth factor receptor the EF-mutant transcription factor is to develop approaches (IGF1R) signaling in Ewing sarcoma has been established. to alter the downstream targets. Creative approaches have The fusion protein positively regulates the expression of been used to screen compounds that are able to negatively IGF1R (29, 30), which has been shown to be necessary for regulate the expression of EWS–FLi1 target genes. A func- EWS–FLi1-mediated transformation of fibroblasts (31). tional screen of a library of 1,040 compounds was per- Ewing sarcoma cells are sensitive to IGF1R inhibition in formed in search for an EWS–FLi1 "off’" signature, and vitro and in vivo (32–35). A phase II trial of R1507, a fully identified cytarabine as a negative modulator of transcrip- human IGF1R-blocking antibody, showed an overall

OF2 Clin Cancer Res; 20(12) June 15, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst April 22, 2014; DOI: 10.1158/1078-0432.CCR-13-0633

New Strategies in Ewing Sarcoma

Insulin-like growth factors Immune clearance IGF1R antibodies IGF1R inhibitors Receptor

TKIs Signaling Lexatumumab NK cells CAR Vorinostat

Figure 1. Current therapeutic RNA interference EWS-FlI1 strategies in Ewing sarcoma. YK-4-279 These include growth factor receptor blockade, intracellular signal inhibition, epigenetic modulation, immune clearance PARP inhibitors enhancement, and manipulation of Mithramycin the EWS–Fli1 transcriptional signature.

Target gene expression/repression

Ewing sarcoma cell growth and survival

© 2014 American Association for Cancer Research

complete response (CR)/partial response (PR) rate of 10% that the most rational combinatorial therapies are used on (36). Similarly, the single-agent activities of the properly selected patients. (IMC-A12) and figitumumab, different human antibodies targeting the same receptor, were reported as 10% and 14%, PARP inhibitors respectively, in patients with refractory Ewing sarcoma (37, PARP inhibitors are an area of growing interest for the 38). A major limitation of these studies was the inability to Ewing sarcoma community. In 2012, a marked sensitivity of identify strategies to select patients most likely to respond to Ewing sarcoma to olaparib, a PARP inhibitor (AZD2281), this therapeutic intervention. was observed in a high-throughput screen of 639 cancer cell Even in responding patients, the majority of responses are lines aimed to detect drug sensitivity patterns as a function short lived; thus, as in other targeted therapies, acquired of genomic features (42). In a different study, sensitivity of resistance to IGF1R blockade has also emerged as a major Ewing sarcoma cells to olaparib was documented both in problem in targeting the IGF1R in Ewing sarcoma, and it is vitro and in tumor xenografts (43). Ewing sarcoma cells were very likely that combination strategies will be needed in the more sensitive to PARP inhibition than prostate cancer cells future to optimize the likelihood of sustained responses. harboring the TMPRSS2-ERG translocation. Remarkably, There is some emerging evidence of mTOR and ERK acti- the combination of temozolamide and olaparib was syn- vation in patients who develop resistance to these therapies ergistic in abrogating progression of Ewing sarcoma xeno- (39). A recent trial combining cixutumumab with temsir- grafts (43). An effect of the EWS–FLI1 fusion transcript in oliumus reported that 35% patients had SD longer than 5 the DNA damage response was suggested more than a months or CR/PR (40). In addition, activation of MAPK and decade ago (44). Interestingly, a high expression of PARP insulin receptor (41) could be involved in resistance to in Ewing sarcoma cells has been reported (45). However, IGF1R inhibition. the exact role of PARP in Ewing sarcoma biology continues Given that the activity of these IGF1R-blocking agents has to be an area of active research. been established, it is imperative that we identify predictors The enthusiasm about these results resulted in a phase II of response as well as mechanisms of acquired resistance so clinical trial of olaparib in recurrent/metastatic Ewing

www.aacrjournals.org Clin Cancer Res; 20(12) June 15, 2014 OF3

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst April 22, 2014; DOI: 10.1158/1078-0432.CCR-13-0633

Arnaldez and Helman

sarcoma following failure of prior chemotherapy. Unfortu- cancer immunotherapy, particularly the positive results seen nately, no CR/PR was seen with 4 of 12 patients achieving after PD-1 blockade in solid tumors (58, 59), have renewed SD at a maximum of 18.4 weeks with a median time to enthusiasm about therapeutic manipulation of the immune progression of 5.7 weeks. Further accrual to this trial was system with the aim of tumor eradication. discontinued (46). Unfortunately, molecular diagnosis was A trial of consolidative immunotherapy for high-risk not a requisite for enrollment, and it is hard to speculate pediatric sarcomas, including Ewing sarcoma, using autol- about the biologic reasons for these results, which could be ogous T cells and dendritic cells pulsed with peptides related not only to lack of the FET–ETS translocation but derived from tumor-specific translocation, was performed also to general lack of predictiveness of current preclinical at the NCI. This approach proved to be feasible and led to models as well as pharmacologic factors. However, it is 31% 5-year overall survival (60). quite possible that other PARP inhibitors, or, combination TRAIL is a member of the TNF superfamily with antitu- therapies, such as with temozolomide, might have a more moral activity secreted primarily by natural killer (NK) cells. auspicious outcome. Ewing sarcoma cells express the TRAIL death receptors, and have been shown to be sensitive to TRAIL-induced caspase- Epigenetic therapies 8–mediated apoptosis in vitro. Tumor progression using Polycomb repressor genes. One of the known down- xenografts and transgene TRAIL expression showed associ- stream targets of EWS–FLi1 is EZH2, which is the catalytic ation of ligand expression with delayed tumor progression subunit of the polycomb repressor gene 2 associated with (61). In a recent phase I trial evaluating lexatumumab, a "stemness" features in tumor cells (47). Expression of EWS– fully human agonistic antibody against TRAIL receptor 2 in FLi1 leads to EZH2 upregulation in mesenchymal stem cells which 4 patients with Ewing sarcoma were enrolled, the (48) and expression of both EZH2 and BIM1 in human agent was well tolerated but no CRs or PRs were observed neural crest cells, although BIM1 is not a direct transcrip- (62). Interestingly, there is potential for synergistic combi- tional target of EWS–FLI1 (49). These findings suggest a nation of immune-based therapies and HDAC inhibitors. rationale for the exploration of the use of the new EZH2 Ewing sarcoma cells treated with vorinostat had increased inhibitors in this tumor (50, 51). sensitivity to TRAIL-induced apoptosis via increased acti- Histone deacetylases. It has been shown that EWS–FLi1 vation of caspase 8 (63). has a transcriptional repressive function (13). One of the Preclinical studies have demonstrated sensitivity of downstream targets of the fusion protein that is required for Ewing sarcoma cells to expanded NK cells in vitro and in oncogenic transformation is NKX2.2 (52). This gene vivo (64). This is congruent with the previous findings that encodes for a transcription factor with both activating and NK cells are able to recognize and destroy Ewing sarcoma repressing domains. NKX2.2 is thought to exert its transcrip- cells by signaling through NKG2D and DNAM-1 receptors tional repression via TLE (transducin-like enhancers of (65). Clinical trials exploring the feasibility of NK-based split)–associated recruitment of histone deacetylases therapy with and without stem cell transplantation in (HDAC). TLE proteins are the homologues of Groucho in patients with high-risk sarcomas, including Ewing sarcoma, humans. They are a family of proteins that act as transcrip- are ongoing (clinicaltrials.gov: NCT01287104 and tional modulators. Expression of individual TLE genes cor- NCT01875601). relates with immature epithelial cells that are progressing Once again, HDAC inhibition has been linked with toward their terminally differentiated state, suggesting a role increased expression of NKG2D ligands in Ewing sarcoma during epithelial differentiation (53). TLE proteins are cells that increased sensitivity to NK cell–mediated cytolysis expressed in Ewing sarcoma and are believed to exert their (66). Ligand upregulation has also been linked to DNA repressive function via recruitment of HDACs. This is a damage—for instance using radiation—(67), all suggesting possible mechanism that could explain preclinical activity that optimal combination or sequential therapies may of HDAC inhibitors in these tumors (54). In vitro HDAC enhance this therapeutic approach. inhibition using vorinostat in the Ewing sarcoma A673 cells Finally, chimeric antigen receptor (CAR)–based therapy led to growth inhibition by abrogation of the transcription- is currently being developed for Ewing sarcoma. Modified T al-repressivefunction (54), which is consistentwith findings cells have shown promising results in hematologic malig- from prior studies in which Ewing sarcoma xenografts nancies (68). Surface receptors expressed in Ewing sarcoma, showed sensitivity to HDAC inhibition (55). Moreover, such as the ganglioside antigen GD2, are being actively combination of 5-aza-20-deoxycytidine, an inhibitor of explored as potential targets for Ewing sarcoma CAR therapy DNA methylation, and an HDAC inhibitor in vitro showed (69, 70). reactivation of tumor suppressor genes and decreased clo- In summary, several recent discoveries are opening a new nogenicity in vitro in Ewing sarcoma cell lines (56). Although era of clinical investigation in Ewing sarcoma. Inhibition of initial clinical trials of this approach have not shown the aberrant fusion protein, PARP inhibition, epigenetic responses (57), this avenue has not been fully explored yet. manipulation, IGF1R blockade, and immune therapies all hold the promise of achieving improved outcomes in Immunotherapy patients with Ewing sarcoma. Immunotherapyshouldbeconsideredasavalidapproach However, despite a sizable number of scientific discov- to Ewing sarcoma therapy. The recent developments in eries in Ewing sarcoma biology, these findings have not led

OF4 Clin Cancer Res; 20(12) June 15, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst April 22, 2014; DOI: 10.1158/1078-0432.CCR-13-0633

New Strategies in Ewing Sarcoma

to a significant improvement in outcomes, particularly in Another issue that may be impeding progress in this rare, patients presenting with metastatic disease. A recent Chil- highly aggressive sarcoma is the paucity of preclinical and dren’s Oncology Group trial demonstrated benefit of inter- clinical models testing rational combinations of signaling val-compressed chemotherapy in patients with localized pathway–targeted agents. With the limited number of disease (7), but as was the case for previous improvements patients available for phase II testing, we may be better in the treatment of Ewing sarcoma, this approach had no served by early testing of combination-targeted agents based impact on the outcome of patients with advanced meta- upon scientific rationale, rather than insisting upon single- static disease. Although the reasons for this remain elusive, agent activity in the phase II setting. some assessment of possible reasons for a disconnect Finally, inadequate correlative studies early on in the between these advances and clinical advances seems development of agents for Ewing sarcoma have markedly warranted. hindered progress. Although tissue acquisition in pediatric The lack of reproducibility of preclinical data, particularly oncology continues to be an area of heated debate, one in oncology, has emerged as a growing concern. Reasons for could argue the dubious ethics of enrolling patients in a trial this could be multifactorial: technical difficulties, lack of in which key biologic questions will remain unanswered. adequate models or even well-meaning experimental bias, In summary, although much biologic insight has been and error. However, it has been argued that the increasing gained in understanding Ewing sarcoma, we must work pressure in the current "publish or perish" culture, difficul- harder to ensure these gains are translated to the clinic. ties in publishing negative results, and scarcity of funding Combination of different approaches in a rational and might have an impact in variable experimental results. creative manner continues to be a challenge for the future. One result of this well-documented phenomenon could To overcome this hurdle, it will be necessary to foster be lack of preclinical rigor required before observations collaboration between very distinct investigative app- are tested in the clinic. For example, many published articles roaches and to appeal to innovative clinical trial designs. using xenograft models demonstrate that a novel inter- If the recent and exciting biologic discoveries can be trans- vention leads to a slowing of tumor growth, but very few lated into effective therapies able to optimize outcomes publications demonstrate actual tumor shrinkage. while minimizing toxicities, we will be able to convey Although tumor shrinkage per se may not predict clinical renewed optimism to patients affected by this deadly tumor efficacy, clearly better preclinical predictors are necessary. In who are in desperate need of new therapeutic approaches. a previous report comparing activity of novel cytotoxic agents in xenografts with activity in phase II studies, the Disclosure of Potential Conflicts of Interest best predictor of clinical activity was when a compound had No potential conflicts of interest were disclosed. activity in at least 33% of multiple histologies in multiple xenograft models (71). As noted above, these data were Authors' Contributions Conception and design: F.I. Arnaldez, L.J. Helman generated using cytotoxic agents, and currently we simply Development of methodology: F.I. Arnaldez do not know what best predicts for clinical activity with Acquisition of data (provided animals, acquired and managed patients, newer "targeted" agents other than a specific activating provided facilities, etc.): L.J. Helman Analysis and interpretation of data (e.g., statistical analysis, biosta- mutation in a kinase predicting for response to a specific tistics, computational analysis): L.J. Helman kinase inhibitor. Unfortunately, no such activating kinase Writing, review, and/or revision of the manuscript: F.I. Arnaldez, mutations have been found in Ewing sarcomas, reinforcing L.J. Helman the point that better predictors for clinical activity are Received August 8, 2013; revised April 4, 2014; accepted April 13, 2014; necessary for this tumor. published OnlineFirst April 22, 2014.

References 1. Esiashvili N, Goodman M, Marcus RB Jr. Changes in incidence and 5. Bernstein ML, Devidas M, Lafreniere D, Souid AK, Meyers PA, Geb- survival of Ewing sarcoma patients over the past 3 decades: surveil- hardt M, et al. Intensive therapy with growth factor support for patients lance epidemiology and end results data. J Pediatr Hematol Oncol with Ewing tumor metastatic at diagnosis: Pediatric Oncology Group/ 2008;30:425–30. Children's Cancer Group Phase II Study 9457–a report from the 2. Rodriguez-Galindo C, Spunt SL, Pappo AS. Treatment of Ewing Children's Oncology Group. J Clin Oncol 2006;24:152–9. sarcoma family of tumors: current status and outlook for the future. 6. Leavey PJ, Mascarenhas L, Marina N, Chen Z, Krailo M, Miser J, Med Pediatr Oncol 2003;40:276–87. et al. Prognostic factors for patients with Ewing sarcoma (EWS) at 3. Grier HE, Krailo MD, Tarbell NJ, Link MP, Fryer CJ, Pritchard DJ, et al. first recurrence following multi-modality therapy: a report from the Addition of ifosfamide and etoposide to standard chemotherapy for Children's Oncology Group. Pediatr Blood Cancer 2008; Ewing's sarcoma and primitive neuroectodermal tumor of bone. N Engl 51:334–8. J Med 2003;348:694–701. 7. Womer RB, West DC, Krailo MD, Dickman PS, Pawel BR, Grier HE, 4. Paulussen M, Craft AW, Lewis I, Hackshaw A, Douglas C, Dunst J, et al. et al. Randomized controlled trial of interval-compressed chemother- Results of the EICESS-92 Study: two randomized trials of Ewing's apy for the treatment of localized Ewing sarcoma: a report from the sarcoma treatment–cyclophosphamide compared with ifosfamide in Children's Oncology Group. J Clin Oncol 2012;30:4148–54. standard-risk patients and assessment of benefit of etoposide added 8. Delattre O, Zucman J, Plougastel B, Desmaze C, Melot T, Peter M, et al. to standard treatment in high-risk patients. J Clin Oncol 2008;26: Gene fusion with an ETS DNA-binding domain caused by chromo- 4385–93. some translocation in human tumours. Nature 1992;359:162–5.

www.aacrjournals.org Clin Cancer Res; 20(12) June 15, 2014 OF5

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst April 22, 2014; DOI: 10.1158/1078-0432.CCR-13-0633

Arnaldez and Helman

9. Kovar H. Dr. Jekyll and Mr. Hyde: the two faces of the FUS/EWS/ sarcoma cell line impairs IGF-1/IGF-1R signalling and reveals TOPK as TAF15 protein family. Sarcoma 2011;2011:837474. a new target. Br J Cancer 2009;101:80–90. 10. Pierron G, Tirode F, Lucchesi C, Reynaud S, Ballet S, Cohen-Gogo S, 30. Cironi L, Riggi N, Provero P, Wolf N, Suva ML, Suva D, et al. IGF1 is a et al. A new subtype of bone sarcoma defined by BCOR-CCNB3 gene common target gene of Ewing's sarcoma fusion proteins in mesen- fusion. Nat Genet 2012;44:461–6. chymal progenitor cells. PLoS One 2008;3:e2634. 11. Sankar S, Lessnick SL. Promiscuous partnerships in Ewing's sarcoma. 31. Toretsky JA, Kalebic T, Blakesley V, LeRoith D, Helman LJ. The insulin- Cancer Genet 2011;204:351–65. like growth factor-I receptor is required for EWS/FLI-1 transformation 12. Hancock JD, Lessnick SL. A transcriptional profiling meta-analysis of fibroblasts. J Biol Chem 1997;272:30822–7. reveals a core EWS-FLI gene expression signature. Cell Cycle 2008; 32. Kolb EA, Gorlick R, Lock R, Carol H, Morton CL, Keir ST, et al. Initial 7:250–6. testing (stage 1) of the IGF-1 receptor inhibitor BMS-754807 by the 13. Sankar S, Bell R, Stephens B, Zhuo R, Sharma S, Bearss DJ, et al. pediatric preclinical testing program. Pediatr Blood Cancer 2011;56: Mechanism and relevance of EWS/FLI-mediated transcriptional 595–603. repression in Ewing sarcoma. Oncogene 2013;32:5089–100. 33. Kang HG, Jenabi JM, Liu XF, Reynolds CP, Triche TJ, Sorensen PH. 14. Lessnick SL, Ladanyi M. Molecular pathogenesis of Ewing sarcoma: Inhibition of the insulin-like growth factor I receptor by epigallocatechin new therapeutic and transcriptional targets. Annu Rev Pathol 2012; gallate blocks proliferation and induces the death of Ewing tumor cells. 7:145–59. Mol Cancer Ther 2010;9:1396–407. 15. Takigami I, Ohno T, Kitade Y, Hara A, Nagano A, Kawai G, et al. 34. Houghton PJ, Morton CL, Gorlick R, Kolb EA, Keir ST, Reynolds CP, Synthetic siRNA targeting the breakpoint of EWS/Fli-1 inhibits growth et al. Initial testing of a (IMC-A12) against IGF-1R of Ewing sarcoma xenografts in a mouse model. Int J Cancer 2011; by the Pediatric Preclinical Testing Program. Pediatr Blood Cancer 128:216–26. 2010;54:921–6. 16. Maksimenko A, Malvy C. Oncogene-targeted antisense oligonucleo- 35. Manara MC, Landuzzi L, Nanni P, Nicoletti G, Zambelli D, Lollini PL, tides for the treatment of Ewing sarcoma. Expert Opin Ther Target et al. Preclinical in vivo study of new insulin-like growth factor-I 2005;9:825–30. receptor–specific inhibitor in Ewing's sarcoma. Clin Cancer Res 2007; 17. Mateo-Lozano S, Gokhale PC, Soldatenkov VA, Dritschilo A, Tirado 13:1322–30. OM, Notario V. Combined transcriptional and translational targeting 36. Pappo AS, Patel SR, Crowley J, Reinke DK, Kuenkele KP, Chawla SP, of EWS/FLI-1 in Ewing's sarcoma. Clin Cancer Res 2006;12: et al. R1507, a monoclonal antibody to the insulin-like growth factor 1 6781–90. receptor, in patients with recurrent or refractory Ewing sarcoma family 18. Ramon AL, Bertrand JR, de Martimprey H, Bernard G, Ponchel G, of tumors: results of a phase II Sarcoma Alliance for Research through Malvy C, et al. siRNA associated with immunonanoparticles directed Collaboration study. J Clin Oncol 2011;29:4541–7. against cd99 antigen improves gene expression inhibition in vivo in 37. Malempati S, Weigel B, Ingle AM, Ahern CH, Carroll JM, Roberts CT, Ewing's sarcoma. J Mol Recognit 2013;26:318–29. et al. Phase I/II trial and pharmacokinetic study of cixutumumab in 19. Toretsky JA, Erkizan V, Levenson A, Abaan OD, Parvin JD, Cripe TP, pediatric patients with refractory solid tumors and Ewing sarcoma: a et al. Oncoprotein EWS-FLI1 activity is enhanced by RNA helicase A. report from the Children's Oncology Group. J Clin Oncol 2012;30: Cancer Res 2006;66:5574–81. 256–62. 20. Erkizan HV, Kong Y, Merchant M, Schlottmann S, Barber-Rotenberg 38. Juergens H, Daw NC, Geoerger B, Ferrari S, Villarroel M, Aerts I, et al. JS, Yuan L, et al. A small molecule blocking oncogenic protein EWS- Preliminary efficacy of the anti-insulin-like growth factor type 1 recep- FLI1 interaction with RNA helicase A inhibits growth of Ewing's sar- tor antibody figitumumab in patients with refractory Ewing sarcoma. J coma. Nat Med 2009;15:750–6. Clin Oncol 2011;29:4534–40. 21. Barber-Rotenberg JS, Selvanathan SP, Kong Y, Erkizan HV, Snyder 39. Subbiah V, Naing A, Brown RE, Chen H, Doyle L, LoRusso P, et al. TM, Hong SP, et al. Single enantiomer of YK-4-279 demonstrates Targeted morphoproteomic profiling of Ewing's sarcoma treated with specificity in targeting the oncogene EWS-FLI1. Oncotarget 2012;3: insulin-like growth factor 1 receptor (IGF1R) inhibitors: response/ 172–82. resistance signatures. PloS One 2011;6:e18424. 22. Stegmaier K, Wong JS, Ross KN, Chow KT, Peck D, Wright RD, et al. 40. Naing A, LoRusso P, Fu S, Hong DS, Anderson P, Benjamin RS, Signature-based small molecule screening identifies cytosine arabi- et al. Insulin growth factor-receptor (IGF-1R) antibody cixutumumab noside as an EWS/FLI modulator in Ewing sarcoma. PLoS Med 2007; combined with the mTOR inhibitor temsirolimus in patients with 4:e122. refractory Ewing's sarcoma family tumors. Clin Cancer Res 23. DuBois SG, Krailo MD, Lessnick SL, Smith R, Chen Z, Marina N, et al. 2012;18:2625–31. Phase II study of intermediate-dose cytarabine in patients with 41. Garofalo C, Mancarella C, Grilli A, Manara MC, Astolfi A, Marino MT, relapsed or refractory Ewing sarcoma: a report from the Children's et al. Identification of common and distinctive mechanisms of resis- Oncology Group. Pediatr Blood Cancer 2009;52:324–7. tance to different anti-IGF-IR agents in Ewing's sarcoma. Mol Endo- 24. Grohar PJ, Griffin LB, Yeung C, Chen QR, Pommier Y, Khanna C, et al. crinol 2012;26:1603–16. Ecteinascidin 743 interferes with the activity of EWS-FLI1 in Ewing 42. Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau sarcoma cells. Neoplasia 2011;13:145–53. KW, et al. Systematic identification of genomic markers of drug 25. Baruchel S, Pappo A, Krailo M, Baker KS, Wu B, Villaluna D, et al. sensitivity in cancer cells. Nature 2012;483:570–5. A phase 2 trial of trabectedin in children with recurrent rhabdomyo- 43. Brenner JC, Feng FY, Han S, Patel S, Goyal SV, Bou-Maroun LM, et al. sarcoma, Ewing sarcoma and non-rhabdomyosarcoma soft tissue PARP-1 inhibition as a targeted strategy to treat Ewing's sarcoma. sarcomas: a report from the Children's Oncology Group. Eur J Cancer Cancer Res 2012;72:1608–13. 2012;48:579–85. 44. Soldatenkov VA, Trofimova IN, Rouzaut A, McDermott F, Dritschilo A, 26. Boro A, Pretre K, Rechfeld F, Thalhammer V, Oesch S, Wachtel M, et al. Notario V. Differential regulation of the response to DNA damage in Small-molecule screen identifies modulators of EWS/FLI1 target gene Ewing's sarcoma cells by ETS1 and EWS/FLI-1. Oncogene 2002;21: expression and cell survival in Ewing's sarcoma. Int J Cancer 2890–5. 2012;131:2153–64. 45. Prasad SC, Thraves PJ, Bhatia KG, Smulson ME, Dritschilo A. 27. Grohar PJ, Woldemichael GM, Griffin LB, Mendoza A, Chen QR, Yeung Enhanced poly(adenosine diphosphate ribose) polymerase activity C, et al. Identification of an inhibitor of the EWS-FLI1 oncogenic and gene expression in Ewing's sarcoma cells. Cancer Res 1990;50: transcription factor by high-throughput screening. J Natl Cancer Inst 38–43. 2011;103:962–78. 46. Choy E, Butrynski J, Harmon D, Morgan J, George S, Wagner A, et al. 28. Kofman S, Perlia CP, Economou SG. Mithramycin in the treatment of Translation of preclinical predictive sensitivity of Ewing sarcoma to metastatic Ewing's sarcoma. Cancer 1973;31:889–93. PARP inhibition: phase II study of olaparib in adult patients with 29. Herrero-Martin D, Osuna D, Ordonez JL, Sevillano V, Martins AS, recurrent/metastatic Ewing sarcoma following failure of prior chemo- Mackintosh C, et al. Stable interference of EWS-FLI1 in an Ewing therapy [abstract]. In: Proceedings of the 104th Annual Meeting of the

OF6 Clin Cancer Res; 20(12) June 15, 2014 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst April 22, 2014; DOI: 10.1158/1078-0432.CCR-13-0633

New Strategies in Ewing Sarcoma

American Association for Cancer Research; 2013 Apr 6–10; Washing- 60. Mackall CL, Rhee EH, Read EJ, Khuu HM, Leitman SF, Bernstein ton, DC: AACR; 2013. D, et al. A pilot study of consolidative immunotherapy in patients 47. Richter GH, Plehm S, Fasan A, Rossler S, Unland R, Bennani-Baiti IM, with high-risk pediatric sarcomas. Clin Cancer Res 2008;14: et al. EZH2 is a mediator of EWS/FLI1 driven tumor growth and 4850–8. metastasis blocking endothelial and neuro-ectodermal differentiation. 61. Picarda G, Lamoureux F, Geffroy L, Delepine P, Montier T, Laud K, Proc Natl Acad Sci U S A 2009;106:5324–9. et al. Preclinical evidence that use of TRAIL in Ewing's sarcoma 48. Riggi N, Suva ML, Suva D, Cironi L, Provero P, Tercier S, et al. EWS-FLI- and osteosarcoma therapy inhibits tumor growth, prevents osteo- 1 expression triggers a Ewing's sarcoma initiation program in primary lysis, and increases animal survival. Clin Cancer Res 2010;16: human mesenchymal stem cells. Cancer Res 2008;68:2176–85. 2363–74. 49. von Levetzow C, Jiang X, Gwye Y, von Levetzow G, Hung L, Cooper A, 62. Merchant MS, Geller JI, Baird K, Chou AJ, Galli S, Charles A, et al. et al. Modeling initiation of Ewing sarcoma in human neural crest cells. Phase I trial and pharmacokinetic study of lexatumumab in pediatric PloS One 2011;6:e19305. patients with solid tumors. J Clin Oncol 2012;30:4141–7. 50. Crea F, Fornaro L, Bocci G, Sun L, Farrar WL, Falcone A, et al. EZH2 63. Sonnemann J, Trommer N, Becker S, Wittig S, Grauel D, Palani CD, inhibition: targeting the crossroad of tumor invasion and angiogenesis. et al. Histone deacetylase inhibitor-mediated sensitization to TRAIL- Cancer Metastasis Rev 2012;31:753–61. induced apoptosis in childhood malignancies is not associated with 51. Crea F, Paolicchi E, Marquez VE, Danesi R. Polycomb genes and upregulation of TRAIL receptor expression, but with potentiated cas- cancer: time for clinical application? Crit Rev Oncol Hematol 2012; pase-8 activation. Cancer Biol Ther 2012;13:417–24. 83:184–93. 64. Cho D, Shook DR, Shimasaki N, Chang YH, Fujisaki H, Campana D. 52. Smith R, Owen LA, Trem DJ, Wong JS, Whangbo JS, Golub TR, et al. Cytotoxicity of activated natural killer cells against pediatric solid Expression profiling of EWS/FLI identifies NKX2.2 as a critical target tumors. Clin Cancer Res 2010;16:3901–9. gene in Ewing's sarcoma. Cancer Cell 2006;9:405–16. 65. Verhoeven DH, de Hooge AS, Mooiman EC, Santos SJ, ten Dam MM, 53. Liu Y, Dehni G, Purcell KJ, Sokolow J, Carcangiu ML, Artavanis- Gelderblom H, et al. NK cells recognize and lyse Ewing sarcoma cells Tsakonas S, et al. Epithelial expression and chromosomal location of through NKG2D and DNAM-1 receptor dependent pathways. Mol human TLE genes: implications for notch signaling and neoplasia. Immunol 2008;45:3917–25. Genomics 1996;31:58–64. 66. Berghuis D, Schilham MW, Vos HI, Santos SJ, Kloess S, Buddingh EP, 54. Owen LA, Kowalewski AA, Lessnick SL. EWS/FLI mediates transcrip- et al. Histone deacetylase inhibitors enhance expression of NKG2D tional repression via NKX2.2 during oncogenic transformation in ligands in Ewing sarcoma and sensitize for natural killer cell-mediated Ewing's sarcoma. PloS One 2008;3:e1965. cytolysis. Clin Sarcoma Res 2012;2:8. 55. Jaboin J, Wild J, Hamidi H, Khanna C, Kim CJ, Robey R, et al. MS-27- 67. Gasser S, Orsulic S, Brown EJ, Raulet DH. The DNA damage pathway 275, an inhibitor of histone deacetylase, has marked in vitro and in vivo regulates innate immune system ligands of the NKG2D receptor. antitumor activity against pediatric solid tumors. Cancer Res 2002; Nature 2005;436:1186–90. 62:6108–15. 68. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. 56. Hurtubise A, Bernstein ML, Momparler RL. Preclinical evaluation of the Chimeric antigen receptor-modified T cells for acute lymphoid leuke- antineoplastic action of 5-aza-20-deoxycytidine and different histone mia. N Engl J Med 2013;368:1509–18. deacetylase inhibitors on human Ewing's sarcoma cells. Cancer Cell 69. Kailayangiri S, Altvater B, Meltzer J, Pscherer S, Luecke A, Dierkes C, Int 2008;8:16. et al. The ganglioside antigen G(D2) is surface-expressed in Ewing 57. Fouladi M, Park JR, Stewart CF, Gilbertson RJ, Schaiquevich P, Sun J, sarcoma and allows for MHC-independent immune targeting. Br J et al. Pediatric phase I trial and pharmacokinetic study of vorinostat: a Cancer 2012;106:1123–33. Children's Oncology Group phase I consortium report. J Clin Oncol 70. Liebsch L, Kailayangiri S, Beck L, Altvater B, Koch R, Dierkes C, et al. 2010;28:3623–9. Ewing sarcoma dissemination and response to T-cell therapy in mice 58. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety assessed by whole-body magnetic resonance imaging. Br J Cancer and tumor responses with lambrolizumab (anti-PD-1) in . N 2013;109:658–66. Engl J Med 2013;369:134–44. 71. Johnson JI, Decker S, Zaharevitz D, Rubinstein LV, Venditti JM, 59. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDer- Schepartz S, et al. Relationships between drug activity in NCI preclin- mott DF, et al. Safety, activity, and immune correlates of anti-PD-1 ical in vitro and in vivo models and early clinical trials. Br J Cancer antibody in cancer. N Engl J Med 2012;366:2443–54. 2001;84:1424–31.

www.aacrjournals.org Clin Cancer Res; 20(12) June 15, 2014 OF7

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst April 22, 2014; DOI: 10.1158/1078-0432.CCR-13-0633

New Strategies in Ewing Sarcoma: Lost in Translation?

Fernanda I. Arnaldez and Lee J. Helman

Clin Cancer Res Published OnlineFirst April 22, 2014.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-13-0633

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2014/05/20/1078-0432.CCR-13-0633. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2014 American Association for Cancer Research.