Introduction

Ewing sarcoma (ES) is an aggressive malignant tumor that occurs in bones and soft tissue and is the second most common bone malignancy after osteosarcoma [1]. Primarily, this disease affects children and adolescents with about 200 patients being diagnosed annually in the USA. For patients with localized and metastatic disease the current standard chemotherapy for ES consists of four to six alternating cycles of VDC/IE (vincristine, doxorubicin, cyclophosphamide/ifosfamide and etoposide) [2, 3•]. The 5-year survival rate for patients with localized disease is about 83%. Unfortunately, patients with metastatic and/or recurrent disease have an overall survival rate of less than 20% [4]. Even those patients who do successfully recover and become long-term survivors often suffer from late effects of their therapy [5, 6, 7•, 8,9,10]. Many of these patients suffer from the development of secondary malignancies including hematopoietic cancers, carcinoma, and other sarcoma like cancers [11]. ES patients also suffer from reduced fertility, renal insufficiency, and cardiomyopathy [8]. Therefore, there is a need to develop treatments that specifically target the underlying biological drivers of this disease. The ES family of tumors (ESFT) is characterized by a unique chromosomal translocation that gives rise to specific gene fusions that involve the EWSR1 gene and ETS transcription factors with the end result being malignant transformation and disease progression. In 85% of ES cases, there is an associated t(11;22) (q24;q12) chromosomal translocation, which leads to the formation of the EWSR1-FLI1 fusion gene [12] whereas in 10–15% of ES cases, there is the EWSR1-ERG fusion gene arising from the t(21;12) (22;12) [12]. The remaining 1–5% of ES cases harbor one of several possible translocations resulting in a fusion gene that contains the EWSR1 gene and a member of the ETS family of transcription factors (Table 1) [1].

Table 1 Summary of the different fusions between non-ETS and ETS genes and their frequency in Ewing sarcoma tumors

Structurally, the EWS and FLI1 components of the fusion protein EWS-FLI1 have unique activity. The c-terminus, comprised of a significant portion of the FLI1 protein, acts as the DNA-binding domain, while EWS, at the N terminus, acts as a transcriptional activator. In general, EWS-FLI1 localizes in the nucleus and binds DNA at GGAA microsatellite repeats to promote transcription of its downstream targets [13, 14]. Notably, the DNA-binding domain is conserved among all 28 members of the ETS family. The transcription program mediated by EWS-ETS leads to malignant transformation, allows cells to maintain a de-differentiated state, and affords them to circumvent toxicity associated with DNA damaging agents [15•, 16, 17]. Preclinical data aimed at understanding the role of EWS-FLI1 in ESFT viability has shown that silencing of EWS-FLI1 expression markedly impairs ES cell growth [18]. Many different approaches have been followed in efforts to develop therapeutic agents that target the biological drivers or malignant phenotype of ES. Here we highlight six classes of experimental agents: (i) DNA-binding agents targeting EWS-FLI1 and protein partners, (ii) agents that reverse transcriptional signature of EWS-FLI1, (iii) kinase inhibitors, (iv) inhibitors of LSD1 (lysine-specific demethylase 1), (v) inhibitors of poly ADP ribose polymerase-1 (PARP-1), and (vi) microtubule inhibitors.

Targeting EWS-FLI1

YK-4-279

The EWS-FLI1 transcriptional complex includes CREB-binding protein, RNA polymerase II, and RNA helicase A (RHA) [19, 20•]. It has been previously reported that RHA increases EWS-FLI1–mediated transcription, suggesting that these protein interactions are important for ES oncogenesis [20•]. EWS-FLI1 binds to RHA in a unique position where if inhibited it would not interfere with any other transcriptional or RNA metabolism proteins (Fig. 1a) [20•]. Based on these experimental discoveries, RHA inhibition is an attractive target for treating ES. Erkizan et al. developed the small molecule YK-4-279 as an inhibitor of RHA: EWS-FLI1 binding. YK-4-279 binds RHA inhibiting its ability to bind and interact with EWS-FLI1 at low micromolar concentrations [21]. They also demonstrated that YK-4-279 treatment inhibits EWS-FLI1 functionality. Luciferase reporter assays were conducted to demonstrate EWS-FLI1 activity at the NR0B1 promoter upon dose dependent YK-4-279 treatment in COS7 cells [21]. YK-4-279 was also shown to induce apoptosis and inhibit ES cell growth [21]. Efficacy studies in ES xenografts displayed significant decreases in tumor size compared with control showing potential use for YK-4-279 in the clinic [21]. Due to poor clinical activity, YK-4-279 is no longer being evaluated for the treatment of ES its clinical derivative TK216 which, is currently being evaluated in patients with relapsedor refractory ES [22•].

Mithramycin

In efforts to discover an EWS-FLI1 inhibitor a 50,000-compound high-throughput screen was conducted at the National Cancer Institute (NCI) in 2011. Cell-based luciferase reporter screens in TC32 ESFT cells identified mithramycin as a potent inhibitor of EWS-FLI1 (Fig. 1b) [23•]. Mithramycin is a tricyclic polyketide that was originally isolated from Streptomyces argillaceus for its antibiotic activity but was later found to have potent anti-tumor activity [24]. Mithramycin was characterized based on its ability to inhibit EWS-FLI1 activity in vitro using microarray expression profiling, qRT-PCR, and immunoblot analysis. In vitro assays displayed mithramycin ability to inhibit expression of downstream EWS-FLI1 targets at the mRNA and protein levels. In vivo studies included xenograft studies where mithramycin suppressed tumor growth in two different ESFT models [23•].

The success of this preclinical study prompted a phase I/II clinical study of mithramycin in children and adults with refractory ES [25•]. Results from the clinical trial showed that mithramycin has a very narrow therapeutic window. At doses relevant to treat and decrease tumor size in these patients, toxicity was prevalent [25•]. The average maximal mithramycin plasma concentration in patients was 17.8 ± 4.6 ng/mL. The average plasma concentrations of mithramycin measured were extremely low compared with the sustained mithramycin concentrations required to suppress EWS–FLI1 transcriptional activity in preclinical studies at ≥ 50 nmol/L [25•]. Patients displayed high levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), hallmarks of severe liver damage [25•]. It has been reported that mithramycin binds DNA specifically at GC sites, inhibiting the transcription factor Sp1 from binding and regulating expression of thousands of genes [26]. There is evidence that the inhibition of Sp1 by mithramycin is a substantial part of its associated toxicity [27]. This data has helped to guide the development of second generation mithramycin analogues that can confer potent inhibition of EWS-FLI1 while bypassing toxicities associated with mithramycin treatment.

Mechanistic studies based on the molecular mode of action of mithramycin have led to the development of novel mithramycin analogues (Fig. 1). MTMSA-Trp and MTMSA-Phe are semi synthetic analogues of mithramycin that have shown in vitro anti-cancer activity similar to mithramycin [28•]. It has also been reported that MTMSA-Trp and MTMSA-Phe bind DNA and physically interact with EWS-FLI1 [29•, 30•]. Further refinement of these mithramycin analogues has led to the development of MTMSA-Phe-Trp and MTMSA-Trp-Trp [31•]. Mitra et al. reported that both MTMSA-Phe-Trp and MTMSA-Trp-Trp have potency similar to mithramycin in ESFT cells [31•]. It was also reported that MTMSA-Phe-Trp and MTMSA-Trp-Trp are more selective for cell lines that express EWS-FLI1 than mithramycin and previously reported analogues [31•]. Further studies are currently ongoing to determine efficacy and pharmacokinetics of other analogues in xenograft mouse models of ES.

Fig. 1
figure 1

Depiction of the mechanism of action of novel treatment approaches for Ewing sarcoma. a YK-4-279 binds RHA inhibiting its interaction with EWS-FLI1 and transcription of target genes. b Mithramycin binds DNA and inhibits EWS-FLI1 mediated transcription. c Trabectedin-binding alkylates DNA distorting its structure and inhibits DNA repair protein activity and transcription factor activity. d LSD1 inhibitors bind LSD1 and prevent its interaction with EWS-FLI1. e, f CDK inhibitors bind cyclin/CDK complexes preventing phosphorylation. g PARP binds DNA and repairs DNA breaks, in the presence of olaparib PARP activity is inhibited rendering the cell vulnerable to consequences of DNA damage. h Eribulin inhibits microtubule growth causing nonproductive tubulin aggregates

Reversing the Transcriptional Signature of EWS-FLI1

Trabectedin

Trabectedin is a natural product that was originally isolated form the sea squirt Ecteinascidia turbinate [32]. Trabectedin binds DNA in the minor groove at GC-rich sequences and alkylates the amino group of guanine at position 2, bending DNA toward the major groove (Fig. 1c) [33]. A potent alkylator, its cytotoxicity comes from its ability to interact with DNA repair pathways. Trabectedin-DNA adducts can trap the transcription-coupled DNA nucleotide excision repair (TC-NER) system as it repairs DNA damage in transcribing cells, this then leads to lethal DNA breaks [34].

Clinically, trabectedin has shown activity in a wide range of malignancies, most notably in leiomyosarcoma and liposarcoma [35]. Previously it has been shown that sarcomas harboring chromosomal translocations are sensitive to trabectedin treatment [36]. Taamma et al. reported a 50% response rate in patients with myxoid liposarcoma which harbors a t(12:16) (q13;p11) chromosomal translocation FUS-CHOP, after treatment with trabectedin [36]. This finding has identified trabectedin as a potential drug of interest for the treatment of ES. Preclinical studies have shown that ES cells are sensitive to trabectedin treatment, and there is mechanistic evidence that trabectedin interferes with EWS-FLI1 activity in vitro, reversing the gene signature of the aberrant transcription factor [37, 38, 39•]. Trabectedin has not shown success in the clinic in ES patients, but there have been efforts to develop analogues that can better target the EWS-FLI1 translocation [40•]. Lurbinectedin, a trabectedin derivative, has shown evidence of activity against EWS-FLI1. This second generation trabectedin analogue causes nuclear redistribution of EWS-FLI1 from the nucleus to the nucleolus, similar to that of the parent compound trabectedin [41•]. Lurbinectedin is currently being tested in a phase 2 clinical trial in select advanced solid tumors [42•].

Inhibitors of LSD1

HCI-2509

Although there have been multiple efforts to target the transcription factor EWS-FLI1, the main oncogenic driver in ES, there has been little success in identifying a potent yet clinically acceptable small molecule inhibitor of the transcription factor. ES has one of the lowest mutation rates of any cancer, and it has emerged as a model system to investigate epigenetic aberrations resulting in oncogenesis [43•, 44, 45•]. Lysine specific demethylase 1 (LSD1) is an enzyme that functions as a histone demethylase and as a transcriptional activator and repressor [46]. LSD1 has been implicated in many malignancies including breast, prostate, bladder, lung, liver, neuroblastoma, amyloid leukemia and colorectal tumors [47,48,49,50,51,52]. In 2011, it was reported that ES expresses unusually high levels of LSD1 [53•, 54]. It has also been demonstrated that overexpression of LSD1 can drive transformation in cells [49, 54, 55]. Previous studies have demonstrated that LSD1 is required for chromosome segregation and downregulation of LSD1 can lead to abnormal centrosome duplication. This significantly impairs nuclear pore complex assembly, leading to an extended telophase [56, 57].

Initially LSD1 inhibition in ES was tested using tranylcypromine [53]. Tranylcypromine is currently prescribed as an anxiolytic and antidepressant. However, because of severe side effects associated with its treatment, its use is limited to treating patients with major depressive disorders [58]. It was shown that tranylcypromine inhibits LSD1 in a non-reversible manner, which translated to decreased cell proliferation at milli-molar concentrations in ES cells, thus supporting further exploration and development of a more potent and specific LSD1 inhibitor [53]. Sankar et al. have been investigating the therapeutic potential of HCI-2509, a non-competitive reversible small molecule inhibitor of LSD1, for the treatment of ES (Fig. 1d) [59•]. Their studies demonstrated that HCI-2509 can reverse the global oncogenic transcriptional program in ES with the use of cell viability assays and RNA-sequencing [59•]. They also demonstrated that HCI-2509 impairs tumorigenesis in vivo. As a single agent HCI-2509 decreased tumor size in comparison to the vehicle group, which led to improved survival of treated animals over 60 days, but these differences were not statistically significant [59•]. Despite this limited efficacy, an analogue of HCI-2509 known as SP-2577 is undergoing phase 1 clinical testing [60].

Kinase Inhibitors

CDK12/13 Inhibitors

One approach to treating ESFT is to target the basic transcriptional machinery with small molecule inhibitors such as THZ1, which is a covalent inhibitor of CDK7/12/13 [61]. The control of gene transcription involves a set of cyclin-dependent kinases (CDKs), including CDK7, CDK8, CDK9, CDK11, CDK12, CDK13, and CDK19 these kinases are essential to transcription, initiation and elongation. Specifically, CDKs phosphorylate RNA polymerase II (RNAPII) at its C-terminal domain (CTD) causing recruitment of transcriptional activators to the transcriptional complex [62]. THZ1 was implicated as a potential therapeutic for treating cancers defined by their high dependency on transcriptional programs for initiating transformation [63]. This was further demonstrated when 1081 cancer cell lines were screened, and ES cell lines were shown to be the most sensitive to THZ1 treatment [64]. Iniguez et al. reported that in ES cell lines THZ1 primarily targets CDK12, a kinase that regulates expression of DNA damage repair genes (Fig. 1e) [64, 65•]. Because of non-specific targeting, the THZ1 analogue THZ1531 was developed and tested in preclinical studies. These studies showed that THZ1531 was highly active in cells expressing EWS-FLI1, specifically showing an increase in proteins involved in double-stranded DNA damage repair. Considering this discovery, they then hypothesized that cells expressing EWS-FLI1 are more vulnerable to THZ1531 and other DNA damage repair inhibitors. ES cell lines have been reported to be highly sensitive to PARP inhibitors [66]. Preclinical combination study results showed strong synergy with THZ1531 and the PARP inhibitor olaparib.

Xenograft studies using THZ1 as a single agent did not cure mice of ES, suggesting that the combination of THZ1 and olaparib could be necessary to achieve complete efficacy [64]. Treatment of tumors with the combination of THZ1 and olaparib demonstrated a marked decrease in tumor size compared with control groups [64].

CDK4/6 Inhibitors

The ES genome is characterized by one of the lowest mutational rates among cancer types, implicating a possibility for epigenetic deregulation as a component for tumor development [44, 45•, 67]. Reports have shown that about 13 to 30% of ES tumors possess deletions in the gene CDKN2A, although these deletions do not appear to be associated with clinical outcome there is still some implication that this could be a target for pharmacological drug development [51, 68]. In ES, the tumor suppressors p16INK4a and p14arf arise from variant transcription start points of the CDKN2A gene, and p16 has been shown to inhibit CDK4 and CDK6–mediated phosphorylation of the RB protein, preventing cell cycle progression [69]. The alteration of p16INK4a in ES has sparked clinical interest in CDK4/6 inhibition. Cyclin D1, a regulatory subunit of CDK4 and CDK6, has also been shown to be a super enhancer in ES [70•] and consequently, in ES cells, the CyclinD1/CDK4 pathway is activated and is required for growth. Kennedy et al. conducted xenograft studies where they demonstrated that CDK4/6 inhibition decreased tumor growth [70•]. These results suggest yet another potential pathway to target oncogenic drivers of ES at the molecular level (Fig. 1f).

Currently, the CDK4/6 inhibitor, abemaciclib is being evaluated in a phase I clinical trial in children and adults with recurrent and refractory ES, neuroblastoma, rhabdomyosarcoma, and osteosarcoma [71]. The main toxicity associated with CDK4/6 inhibition is hematologic in nature, and there is growing interest in combining CDK4/6 inhibitors with cytotoxic chemotherapeutics that have non-overlapping toxicities [72]. There is also interest in combining CDK4/6 inhibitors with MEK, mTOR, and IGF-1R inhibitors, exploiting multiple pathways focusing on targets that can influence tumor growth in ES [73,74,75].

PARP-1 Inhibitors

Olaparib

Mechanistically, poly adenosine diphosphate ribose polymerase (PARP) is an enzyme that has been shown to both drive transcription and to accelerate base excision repair (Fig. 1g) [76,77,78]. Initial interest in PARP1 inhibitors have shown promising activity particularly in BRCA-mutant cancers defective in homologous repair (HR), in which they have demonstrated replication fork stalling and subsequent synthetic lethal cell death [79,80,81]. PARP inhibitors have also recently been of interest for the treatment of ES. It has been reported that ES cells express high levels of PARP mRNA and protein which translates to high PARP activity related to an increase in copy number compared with other cancers [82]. Increased PARP activity has led to the report that inhibition of PARP sensitizes ES cells to ionizing radiation [82]. In more recent studies Brenner et al. showed increased sensitivity of three ES cell lines to the PARP inhibitor olaparib, as opposed to an osteosarcoma and rhabdomyosarcoma cell line [83•]. In this study, however, olaparib was not effective as a single agent in xenograft experiments. They subsequently combined olaparib with temozolomide, a DNA alkylating agent, results displayed a marked decrease in tumor size of the xenograft models. These data represent the potential for elucidating synthetic lethality in ES cells in the presence of a PARP inhibitor and a DNA damaging agent [83•]. In 2014, a phase II clinical study was conducted evaluating the antitumor activity of olaparib, as a monotherapy in patients with refractory ES following failure of standard chemotherapy [84•]. This study concluded that olaparib administration was safe and well tolerated in patients. However, as a monotherapy, there were no significant responses or durable disease control [84•]. Currently a phase I study of olaparib and temozolomide for the treatment of ES is ongoing [85] .

Microtubule Inhibitors

Eribulin

Microtubules help support organelles, transport molecules, and give shape to the cell. The microtubule inhibitor eribulin inhibits polymerization of tubulin subunits by preventing lengthening and shortening of microtubules during cellular division (Fig. 1h) [86, 87]. When these unstable polymers of tubulin aggregate the result is apoptosis [88, 89]. Eribulin is approved by the FDA for treating metastatic breast cancer and may have promising activity in other cancers [90]. In ES, EWS-FLI1 drives expression of proteins that regulate microtubule stability, making this an attractive drug target. In pre-clinical studies, testing of eribulin in ES cell lines demonstrated Bcl-2 induced apoptosis [91]. Additionally, eribulin is FDA-approved for adult patients with liposarcoma who previously received an anthracycline [92]. An ongoing phase 2 trial is evaluating eribulin in patients with relapsed/refractory rhabdomyosarcoma and ES [93]. Another phase 1/2 clinical trial is evaluating the combination of eribulin with irinotecan in children with refractory and recurrent rhabdomyosarcoma and ES as well [94]. Overall, eribulin has shown strong preclinical and clinical results in ES, thus giving it great potential as either a monotherapy and in combination with another approved chemotherapeutic.

Conclusion

ES treatment relies on combinations of surgery, radiation, and traditional chemotherapeutic regiments. In many ES patients, these conventional treatment approaches are not enough to remedy this aggressive cancer. Additionally, ES patients in remission must deal with considerable acute and long-term toxicities associated with these therapies. Improving the outcomes for patients with ES will require development of targeted therapies. Therefore, a thorough understanding of the activity, translation, and verification of novel agents is vital in targeting biologically relevant drivers of ES. This is important for clinical development of successful targeted therapies. In this review, we focused on several approaches targeting specific pathways that play a part in ESFT growth in patients. Targeting the EWS–FLI1 transcription factor directly is an approach that has shown promising results but has also been met with many challenges. A major challenge will be to discover a pharmacologic inhibitor that selectively acts to disrupt the function of EWS-FLI1. Alternatively, blocking important pathways, inhibiting the downstream gene signature of EWS-FLI1 and exploiting PARP inhibitor sensitivities have shown promising preclinical results that have yet to be translated in the clinic. Continued efforts toward developing novel therapeutics targeting specific molecular abnormalities in ES are currently ongoing in efforts to improve survival outcomes for these patients.