Immunotherapy
Previous studies in adult malignancies have suggested that widespread expression of programed death ligand-1 (PD-L1) in malignant cells, high mutational tumor burden, and extensive infiltration of tumors with CD8+ T cells were all associated with responses to immune checkpoint blockade.67Although results have varied between studies, Machado et al identified PD-L1 expression in tumor cells in 19% of Ewing sarcoma samples in the largest series to date.68 However, Ewing sarcoma has a low mutational tumor burden when compared to carcinomas or melanoma.69–72 Further, Ewing sarcomas typically have only a low level of infiltrating T cells, identified in only 15% of tumor samples in the Machado series.68 These laboratory findings suggest that Ewing sarcoma would be a relatively “cold” tumor in terms of responding to immunotherapy, and indeed two cooperative group clinical trials reported to date are consistent with this impression. Specifically, no responses have been reported in 13 patients treated with the anti-PD-1 antibody pembrolizumab73 or in ten patients treated with the anti-PD-1 antibody nivolumab.74 While it is hoped that combination immunotherapy regimens now under investigation will make sarcomas more “hot” and therefore responsive,75 at present there is little evidence that checkpoint inhibitors should routinely be used as monotherapy for relapsed Ewing sarcoma.
An important caveat is the use of an innovative immunotherapy approach for Ewing sarcoma called Vigil.76,77 This novel strategy involves administration of a vaccine that comprised autologous tumor cells to provide patient-specific tumor antigens to provoke an anti-tumor response. These cells are transfected with the rhGMCSF transgene and the RNAibi-shRNAfurin in order to recruit and activate dendritic cells while reducing local immune tolerance through the blockade of furin-mediated activation of endogenous TGF-beta1 and 2. An early trial reported a 1-year survival of 73% for patients with relapsed Ewing sarcoma treated with Vigil compared to 23% of historical controls treated with conventional chemotherapy.78 Given the low toxicity of Vigil, a randomized Phase III trial is now underway that combines Vigil with TEM/IRN and compares this group with patients treated with TEM/IRN alone [NCT03495921]. This trial represents one of the few Phase III studies for relapsed Ewing sarcoma, and hopefully accrual of patients from both pediatric and adult sarcoma centers will allow for meaningful conclusions.
Should relapsed patients be treated with high-dose chemotherapy and autologous stem cell transplantation?
Several retrospective studies have suggested that treatment with myeloablative doses of alkylators followed by autologous stem cell transplantation may improve outcomes in patients with recurrent Ewing sarcoma.79–81 Most patients in these studies first received conventional-dose chemotherapy, which was then followed by high-dose busulfan and melphalan. To date, no prospective randomized studies have been performed, and the difficulty in identifying a suitable control population has made interpretation of results more complicated. For example, patients who receive high-dose chemotherapy generally have recurrent tumor that is responsive to typical salvage therapy. At best, this describes only about half of relapsed patients,80 with the majority being those with initially localized disease who often have longer survival than relapsed patients who had metastases at diagnosis. In addition, patients must remain progression-free until high-dose chemotherapy is administered, must have adequate stem cells collected, and must have no comorbidities or organ dysfunction that would preclude such intensive treatment. Finally, patients must be willing to undergo this intensive treatment, which is not always the case given that their prognosis still remains unfavorable, despite the prolonged therapy they have already received.
Taken together, the data would suggest that certain patients with favorable features at relapse may possibly have prolonged PFS with high-dose therapy. In a different clinical context, newly diagnosed patients with localized higher-risk tumors were randomized to receive either myeloablative busulfan/melpahalan with autologous stem cells or continuation of standard chemotherapy.82Although patients receiving high-dose therapy had superior outcomes, it should be emphasized that the treatment backbone used in European studies of newly diagnosed patients differs from that used in North America,83 and the implications of this study for relapsed patients are unclear. Moreover, a study incorporating high-dose chemotherapy in patients with responsive recurrent disease was not able to show it to be an independent variable influencing post-relapse survival.10 The analysis was complicated by the fact that high-dose chemotherapy was reserved for patients achieving a complete or partial response to IFOS, no prior history of myeloablative therapy, and willingness to undergo high-intensity treatment.10 As such, only 20/107 evaluable patients underwent treatment.10 This study illustrates some of the complexities of designing a clinical trial to rigorously demonstrate a role for high-dose therapy and autologous stem cell transplantation in the relapse setting. It is clear that this strategy is not well suited for all relapsed patients, and it remains an intensive therapy for which the unequivocal benefits have not yet been established.
What is the role of genetic testing of tumor tissue for actionable mutations in patients with recurrent Ewing sarcoma?
Apart from the characteristic EWSR1 translocations that characterize this tumor, recurring genetic changes in Ewing sarcoma are relatively infrequent. The most common mutations occur in genes such as STAG2, CDKN2A, and TP53, which have not been easily targetable.69–71 Although potentially actionable mutations have been reported with Ewing sarcoma, these are uncommon.84 Therefore, the likelihood of identifying a molecular change that will drive effective therapy is low but not zero. Ongoing translational studies that prospectively collect data on patients with relapsed Ewing sarcoma will be helpful to further characterize the genetic changes that may occur in these patients, especially given the potential of new targeted agents being developed. However, extensive molecular testing outside the context of a clinical trial is expensive, and decisions regarding genetic analysis should be individualized given that the cost/benefit relationship of such testing has not been clarified.
CONCLUSION
The outcome for patients with recurrent Ewing sarcoma remains poor, and the standard approach to their management has not yet been established. Many patients may initially benefit from salvage chemotherapy in terms of reducing symptoms and prolonging time to further progression, but consistent cures remain elusive. Knowledge of prognostic factors that affect survival of these patients may help guide therapy decisions. Enrollment on clinical trials should strongly be considered when feasible, as a variety of mechanistically novel Phase I to Phase III studies are currently underway and represent the best way to better understand which treatments may be beneficial in the future. In this regard, close cooperation between pediatric and adult oncology centers, as well as between continents, will help drive advances for this rare disease of adolescence and young adulthood.
Disclosure
The authors report no conflicts of interest in this work.
David Van Mater, Lars Wagner
Department of Pediatrics, Division of Hematology/Oncology, Duke University, Durham, NC, USA.
References
1. Womer RB, West DC, Krailo MD, et al. Randomized controlled trial of interval-compressed chemotherapy for the treatment of localized Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol. 2012;30(33):4148–4154. doi:10.1200/JCO.2011.41.5703
2. Juergens C, Weston C, Lewis I, et al. Safety assessment of intensive induction with vincristine, ifosfamide, doxorubicin, and etoposide (VIDE) in the treatment of Ewing tumors in the EURO-E.W.I.N.G. 99 clinical trial. Pediatr Blood Cancer. 2006;47(1):22–29. doi:10.1002/pbc.20820
3. Wagner MJ, Gopalakrishnan V, Ravi V, et al. Vincristine, ifosfamide, and doxorubicin for initial treatment of Ewing sarcoma in adults. Oncologist. 2017;22(10):1271–1277. doi:10.1634/theoncologist.2016-0464
4. Gaspar N, Hawkins DS, Dirksen U, et al. Ewing sarcoma: current management and future approaches through collaboration. J Clin Oncol. 2015;33(27):3036–3046. doi:10.1200/JCO.2014.59.5256
5. Leavey PJ, Mascarenhas L, Marina N, et al. Prognostic factors for patients with Ewing sarcoma (EWS) at first recurrence following multi-modality therapy: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2008;51(3):334–338. doi:10.1002/pbc.21618
6. Stahl M, Ranft A, Paulussen M, et al. Risk of recurrence and survival after relapse in patients with Ewing sarcoma. Pediatr Blood Cancer. 2011;57(4):549–553. doi:10.1002/pbc.23040
7. Bacci G, Longhi A, Ferrari S, et al. Pattern of relapse in 290 patients with nonmetastatic Ewing’s sarcoma family tumors treated at a single institution with adjuvant and neoadjuvant chemotherapy between 1972 and 1999. Eur J Surg Oncol. 2006;32(9):974–979. doi:10.1016/j.ejso.2006.01.023
8. Rodriguez-Galindo C, Billups CA, Kun LE, et al. Survival after recurrence of Ewing tumors: the St Jude Children’s Research Hospital experience, 1979–1999. Cancer. 2002;94(2):561–569. doi:10.1002/cncr.10192
9. Shankar AG, Ashley S, Craft AW, Pinkerton CR. Outcome after relapse in an unselected cohort of children and adolescents with Ewing sarcoma. Med Pediatr Oncol. 2003;40(3):141–147. doi:10.1002/mpo.10248
10. Ferrari S, Luksch R, Hall KS, et al. Post-relapse survival in patients with Ewing sarcoma. Pediatr Blood Cancer. 2015;62(6):994–999. doi:10.1002/pbc.25388
11. Wasilewski-Masker K, Liu Q, Yasui Y, et al. Late recurrence in pediatric cancer: a report from the Childhood Cancer Survivor Study. J Natl Cancer Inst. 2009;101(24):1709–1720. doi:10.1093/jnci/djp417
12. Heinemann M, Ranft A, Langer T, et al. Recurrence of Ewing sarcoma: is detection by imaging follow-up protocol associated with survival advantage? Pediatr Blood Cancer. 2018;65(7):e27011. doi:10.1002/pbc.27011
13. Palmerini E, Jones RL, Setola E, et al. Irinotecan and temozolomide in recurrent Ewing sarcoma: an analysis in 51 adult and pediatric patients. Acta Oncol. 2018;57(7):958–964. doi: 10.1080/0284186X.2018.1449250.
14. Scobioala S, Ranft A, Wolters H, et al. Impact of whole lung irradiation on survival outcome in patients with lung relapsed Ewing sarcoma. Int J Radiat Oncol Biol Phys. 2018;102(3):584–592. doi:10.1016/j.ijrobp.2018.06.032
15. Ferrari S, Del Prever AB, Palmerini E, et al. Response to high-dose ifosfamide in patients with advanced/recurrent Ewing sarcoma. Pediatr Blood Cancer. 2009;52(5):581–584. doi:10.1002/pbc.21917
16. Hunold A, Weddeling N, Paulussen M, Ranft A, Liebscher C, Jurgens H. Topotecan and cyclophosphamide in patients with refractory or relapsed Ewing tumors. Pediatr Blood Cancer. 2006;47(6):795–800. doi:10.1002/pbc.20719
17. Saylors RL 3rd, Stine KC, Sullivan J, et al. Cyclophosphamide plus topotecan in children with recurrent or refractory solid tumors: a Pediatric Oncology Group phase II study. J Clin Oncol. 2001;19(15):3463–3469. doi:10.1200/JCO.2001.19.15.3463
18. Farhat R, Raad R, Khoury NJ, et al. Cyclophosphamide and topotecan as first-line salvage therapy in patients with relapsed ewing sarcoma at a single institution. J Pediatr Hematol Oncol. 2013;35(5):356–360. doi:10.1097/MPH.0b013e318270a343
19. Wagner LM, McAllister N, Goldsby RE, et al. Temozolomide and intravenous irinotecan for treatment of advanced Ewing sarcoma. Pediatr Blood Cancer. 2007;48(2):132–139. doi:10.1002/pbc.20697
20. Casey DA, Wexler LH, Merchant MS, et al. Irinotecan and temozolomide for Ewing sarcoma: the Memorial Sloan-Kettering experience. Pediatr Blood Cancer. 2009;53(6):1029–1034. doi:10.1002/pbc.22206
21. Anderson P, Kopp L, Anderson N, et al. Novel bone cancer drugs: investigational agents and control paradigms for primary bone sarcomas (Ewing’s sarcoma and osteosarcoma). Expert Opin Investig Drugs. 2008;17(11):1703–1715. doi:10.1517/13543784.17.11.1703
22. Raciborska A, Bilska K, Drabko K, et al. Vincristine, irinotecan, and temozolomide in patients with relapsed and refractory Ewing sarcoma. Pediatr Blood Cancer. 2013;60(10):1621–1625. doi:10.1002/pbc.24621
23. Kurucu N, Sari N, Ilhan IE. Irinotecan and temozolamide treatment for relapsed Ewing sarcoma: a single-center experience and review of the literature. Pediatr Hematol Oncol. 2015;32(1):50–59. doi:10.3109/08880018.2014.954070
24. Buyukkapu Bay S, Kebudi R, Gorgun O, Zulfikar B, Darendeliler E, Cakir FB. Vincristine, irinotecan, and temozolomide treatment for refractory/relapsed pediatric solid tumors: a single center experience. J Oncol Pharm Pract. 2018:1078155218790798. Epub 2018 Aug 6.
25. Fox E, Patel S, Wathen JK, et al. Phase II study of sequential gemcitabine followed by docetaxel for recurrent Ewing sarcoma, osteosarcoma, or unresectable or locally recurrent chondrosarcoma: results of Sarcoma Alliance for Research Through Collaboration Study 003. Oncologist. 2012;17(3):321. doi:10.1634/theoncologist.2010-0265
26. Mora J, Cruz CO, Parareda A, de Torres C. Treatment of relapsed/refractory pediatric sarcomas with gemcitabine and docetaxel. J Pediatr Hematol Oncol. 2009;31(10):723–729. doi:10.1097/MPH.0b013e3181b2598c
27. Tanaka K, Joyama S, Chuman H, et al. Feasibility and efficacy of gemcitabine and docetaxel combination chemotherapy for bone and soft tissue sarcomas: multi-institutional retrospective analysis of 134 patients. World J Surg Oncol. 2016;14(1):306. doi:10.1186/s12957-016-1059-2
28. Wagner L. Camptothecin-based regimens for treatment of ewing sarcoma: past studies and future directions. Sarcoma. 2011;2011:957957. doi:10.1155/2011/957957
29. Wagner LM, Perentesis JP, Reid JM, et al. Phase I trial of two schedules of vincristine, oral irinotecan, and temozolomide (VOIT) for children with relapsed or refractory solid tumors: a Children’s Oncology Group phase I consortium study. Pediatr Blood Cancer. 2010;54(4):538–545. doi:10.1002/pbc.22407
30. Pappo AS, Lyden E, Breitfeld P, et al. Two consecutive phase II window trials of irinotecan alone or in combination with vincristine for the treatment of metastatic rhabdomyosarcoma: the Children’s Oncology Group. J Clin Oncol. 2007;25(4):362–369. doi:10.1200/JCO.2006.07.1720
31. Mascarenhas L, Lyden ER, Breitfeld PP, et al. Randomized phase II window trial of two schedules of irinotecan with vincristine in patients with first relapse or progression of rhabdomyosarcoma: a report from the Children’s Oncology Group. J Clin Oncol. 2010;28(30):4658–4663. doi:10.1200/JCO.2010.29.7390
32. Wagner LM. Oral irinotecan for treatment of pediatric solid tumors: ready for prime time? Pediatr Blood Cancer. 2010;54(5):661–662. doi:10.1002/pbc.22410
33. Wagner LM, Crews KR, Stewart CF, et al. Reducing irinotecan-associated diarrhea in children. Pediatr Blood Cancer. 2008;50(2):201–207. doi:10.1002/pbc.21280
34. Wagner L, Turpin B, Nagarajan R, Weiss B, Cripe T, Geller J. Pilot study of vincristine, oral irinotecan, and temozolomide (VOIT regimen) combined with bevacizumab in pediatric patients with recurrent solid tumors or brain tumors. Pediatr Blood Cancer. 2013;60(9):1447–1451. doi:10.1002/pbc.24547
35. Bagatell R, Norris R, Ingle AM, et al. Phase 1 trial of temsirolimus in combination with irinotecan and temozolomide in children, adolescents and young adults with relapsed or refractory solid tumors: a Children’s Oncology Group Study. Pediatr Blood Cancer. 2014;61(5):833–839. doi:10.1002/pbc.24874
36. van Maldegem AM, Benson C, Rutkowski P, et al. Etoposide and carbo-or cisplatin combination therapy in refractory or relapsed Ewing sarcoma: a large retrospective study. Pediatr Blood Cancer. 2015;62(1):40–44. doi:10.1002/pbc.25230
37. Van Winkle P, Angiolillo A, Krailo M, et al. Ifosfamide, carboplatin, and etoposide (ICE) reinduction chemotherapy in a large cohort of children and adolescents with recurrent/refractory sarcoma: the Children’s Cancer Group (CCG) experience. Pediatr Blood Cancer. 2005;44(4):338–347. doi:10.1002/pbc.20227
38. Podda MG, Luksch R, Puma N, et al. Oral etoposide in relapsed or refractory Ewing sarcoma: a monoinstitutional experience in children and adolescents. Tumori. 2016;102(1):84–88. doi:10.5301/tj.5000419
39. Kolb EA, Gorlick R, Reynolds CP, et al. Initial testing (stage 1) of eribulin, a novel tubulin binding agent, by the pediatric preclinical testing program. Pediatr Blood Cancer. 2013;60(8):1325–1332. doi:10.1002/pbc.24517
40. Schafer ES, Rau RE, Berg S, et al. A phase 1 study of eribulin mesylate (E7389), a novel microtubule-targeting chemotherapeutic agent, in children with refractory or recurrent solid tumors: a Children’s Oncology Group Phase 1 Consortium study (ADVL1314). Pediatr Blood Cancer. 2018;65(8):e27066. doi:10.1002/pbc.27066
41. Moreno L, Casanova M, Chisholm JC, et al. Phase I results of a phase I/II study of weekly nab-paclitaxel in paediatric patients with recurrent/refractory solid tumours: a collaboration with innovative therapies for children with cancer. Eur J Cancer. 2018;100:27–34. doi:10.1016/j.ejca.2018.05.002
42. Wagner LM, Yin H, Eaves D, Currier M, Cripe TP. Preclinical evaluation of nanoparticle albumin-bound paclitaxel for treatment of pediatric bone sarcoma. Pediatr Blood Cancer. 2014;61(11):2096–2098. doi:10.1002/pbc.25062
43. Kang MH, Wang J, Makena MR, et al. Activity of MM-398, nanoliposomal irinotecan (nal-IRI), in Ewing’s family tumor xenografts is associated with high exposure of tumor to drug and high SLFN11 expression. Clin Cancer Res. 2015;21(5):1139–1150. doi:10.1158/1078-0432.CCR-14-1882
44. Kolb EA, Gorlick R. Development of IGF-IR inhibitors in pediatric sarcomas. Curr Oncol Rep. 2009;11(4):307–313.
45. Pappo AS, Patel SR, Crowley J, et al. R1507, a monoclonal antibody to the insulin-like growth factor 1 receptor, in patients with recurrent or refractory Ewing sarcoma family of tumors: results of a phase II Sarcoma Alliance for Research Through Collaboration Study. J Clin Oncol. 2011;29(34):4541–4547. doi:10.1200/JCO.2010.34.0000
46. Juergens H, Daw NC, Geoerger B, et al. Preliminary efficacy of the anti-insulin-like growth factor type 1 receptor antibody figitumumab in patients with refractory Ewing sarcoma. J Clin Oncol. 2011;29(34):4534–4540. doi:10.1200/JCO.2010.33.0670
47. Malempati S, Weigel B, Ingle AM, et al. Phase I/II trial and pharmacokinetic study of cixutumumab in pediatric patients with refractory solid tumors and Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol. 2012;30(3):256–262. doi:10.1200/JCO.2011.37.4355
48. Tap WD, Demetri G, Barnette P, et al. Phase II study of ganitumab, a fully human anti-type-1 insulin-like growth factor receptor antibody, in patients with metastatic Ewing family tumors or desmoplastic small round cell tumors. J Clin Oncol. 2012;30(15):1849–1856. doi:10.1200/JCO.2011.37.2359
49. Naing A, LoRusso P, Fu S, et al. Insulin growth factor-receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with refractory Ewing’s sarcoma family tumors. Clin Cancer Res. 2012;18(9):2625–2631. doi:10.1158/1078-0432.CCR-12-0061
50. Schwartz GK, Tap WD, Qin LX, et al. Cixutumumab and temsirolimus for patients with bone and soft-tissue sarcoma: a multicentre, open-label, phase 2 trial. Lancet Oncol. 2013;14(4):371–382. doi:10.1016/S1470-2045(13)70049-4
51. Wagner LM, Fouladi M, Ahmed A, et al. Phase II study of cixutumumab in combination with temsirolimus in pediatric patients and young adults with recurrent or refractory sarcoma: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2015;62(3):440–444. doi:10.1002/pbc.25334
52. Richter GH, Fasan A, Hauer K, et al. G-Protein coupled receptor 64 promotes invasiveness and metastasis in Ewing sarcomas through PGF and MMP1. J Pathol. 2013;230(1):70–81. doi:10.1002/path.4170
53. Nielsen DL, Sengelov L. Inhibition of placenta growth factor with TB-403: a novel antiangiogenic cancer therapy. Expert Opin Biol Ther. 2012;12(6):795–804. doi:10.1517/14712598.2012.679655
54. Attia S, Bolejack V, Ganjoo KN, et al. A phase II trial of regorafenib (REGO) in patients (pts) with advanced Ewing sarcoma and related tumors (EWS) of soft tissue and bone: SARC024 trial results [abstract]. J Clin Oncol. 2017;35:11005. doi:10.1200/JCO.2017.35.15_suppl.11005
55. Italiano A, Penel N, Toulmonde M, et al. Cabozantinib in patients with advanced osteosarcomas and Ewing sarcomas: a French Sarcoma Group (FSG)/US National Cancer Institute phase II collaborative study. Poster presented at: European Society for Medical Oncology; October 19–23; 2018; Munich, Germany.
56. Fleuren ED, Roeffen MH, Leenders WP, et al. Expression and clinical relevance of MET and ALK in Ewing sarcomas. Int J Cancer. 2013;133(2):427–436. doi:10.1002/ijc.28047
57. van Maldegem AM, Bovee JV, Peterse EF, Hogendoorn PC, Gelderblom H. Ewing sarcoma: the clinical relevance of the insulin-like growth factor 1 and the poly-ADP-ribose-polymerase pathway. Eur J Cancer. 2016;53:171–180. doi:10.1016/j.ejca.2015.09.009
58. Choy E, Butrynski JE, Harmon DC, et al. Phase II study of olaparib in patients with refractory Ewing sarcoma following failure of standard chemotherapy. BMC Cancer. 2014;14:813. doi:10.1186/1471-2407-14-813
59. Stewart E, Goshorn R, Bradley C, et al. Targeting the DNA repair pathway in Ewing sarcoma. Cell Rep. 2014;9(3):829–841. doi:10.1016/j.celrep.2014.09.028
60. Zollner SK, Selvanathan SP, Graham GT, et al. Inhibition of the oncogenic fusion protein EWS-FLI1 causes G2-M cell cycle arrest and enhanced vincristine sensitivity in Ewing’s sarcoma. Sci Signal. 2017;10:499. doi:10.1126/scisignal.aam8429
61. Gollavilli PN, Pawar A, Wilder-Romans K, et al. EWS/ETS-driven Ewing sarcoma requires BET bromodomain proteins. Cancer Res. 2018;78(16):4760–4773. doi:10.1158/0008-5472.CAN-18-0484
62. Mancarella C, Pasello M, Ventura S, et al. Insulin-like growth factor 2 mRNA-binding protein 3 is a novel post-transcriptional regulator of Ewing sarcoma malignancy. Clin Cancer Res. 2018;24(15):3704–3716. doi:10.1158/1078-0432.CCR-17-2602
63. Jacques C, Lamoureux F, Baud’huin M, et al. Targeting the epigenetic readers in Ewing sarcoma inhibits the oncogenic transcription factor EWS/Fli1. Oncotarget. 2016;7(17):24125–24140. doi:10.18632/oncotarget.8214
64. Loganathan SN, Tang N, Fleming JT, et al. BET bromodomain inhibitors suppress EWS-FLI1-dependent transcription and the IGF1 autocrine mechanism in Ewing sarcoma. Oncotarget. 2016;7(28):43504–43517. doi:10.18632/oncotarget.9762
65. Hensel T, Giorgi C, Schmidt O, et al. Targeting the EWS-ETS transcriptional program by BET bromodomain inhibition in Ewing sarcoma. Oncotarget. 2016;7(2):1451–1463. doi:10.18632/oncotarget.6385
66. Theisen ER, Pishas KI, Saund RS, Lessnick SL. Therapeutic opportunities in Ewing sarcoma: EWS-FLI inhibition via LSD1 targeting. Oncotarget. 2016;7(14):17616–17630. doi:10.18632/oncotarget.7124
67. Tong M, Wang J, He W, et al. Predictive biomarkers for tumor immune checkpoint blockade. Cancer Manag Res. 2018;10:4501–4507. doi:10.2147/CMAR.S179680
68. Machado I, Lopez-Guerrero JA, Scotlandi K, Picci P, Llombart-Bosch A. Immunohistochemical analysis and prognostic significance of PD-L1, PD-1, and CD8+tumor-infiltrating lymphocytes in Ewing’s sarcoma family of tumors (ESFT). Virchows Arch. 2018;472(5):815–824. doi:10.1007/s00428-018-2316-2
69. Crompton BD, Stewart C, Taylor-Weiner A, et al. The genomic landscape of pediatric Ewing sarcoma. Cancer Discov. 2014;4(11):1326–1341. doi:10.1158/2159-8290.CD-13-1037
70. Tirode F, Surdez D, Ma X, et al. Genomic landscape of Ewing sarcoma defines an aggressive subtype with co-association of STAG2 and TP53 mutations. Cancer Discov. 2014;4(11):1342–1353. doi:10.1158/2159-8290.CD-14-0622
71. Brohl AS, Solomon DA, Chang W, et al. The genomic landscape of the Ewing sarcoma family of tumors reveals recurrent STAG2 mutation. PLoS Genet. 2014;10(7):e1004475. doi:10.1371/journal.pgen.1004541
72. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome landscapes. Science. 2013;339(6127):1546–1558. doi:10.1126/science.1235122
73. Tawbi HA, Burgess M, Bolejack V, et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 2017;18(11):1493–1501. doi:10.1016/S1470-2045(17)30624-1
74. Davis KL, Fox E, Reid JM, et al. ADVL1412: initial results of a phase I/II study of nivolumab and ipilimumab in pediatric patients with relapsed/refractory solid tumors – A COG study [abstract]. J Clin Oncol. 2017;35:10526. doi:10.1200/JCO.2017.35.15_suppl.10526
75. D’Angelo SP, Mahoney MR, Van Tine BA, et al. Nivolumab with or without ipilimumab treatment for metastatic sarcoma (Alliance A091401): two open-label, non-comparative, randomised, phase 2 trials. Lancet Oncol. 2018;19(3):416–426. doi:10.1016/S1470-2045(18)30006-8
76. Ghisoli M, Barve M, Schneider R, et al. Pilot trial of FANG immunotherapy in Ewing’s sarcoma. Mol Ther. 2015;23(6):1103–1109. doi:10.1038/mt.2015.43
77. Rao DD, Jay C, Wang Z, et al. Preclinical Justification of pbi-shRNA EWS/FLI1 lipoplex (LPX) treatment for Ewing’s sarcoma. Mol Ther. 2016;24(8):1412–1422. doi:10.1038/mt.2016.93
78. Ghisoli M, Barve M, Mennel R, et al. Three-year follow up of GMCSF/bi-shRNA(furin) DNA-transfected autologous tumor immunotherapy (Vigil) in metastatic advanced Ewing’s sarcoma. Mol Ther. 2016;24(8):1478–1483. doi:10.1038/mt.2016.86
79. Rasper M, Jabar S, Ranft A, Jurgens H, Amler S, Dirksen U. The value of high-dose chemotherapy in patients with first relapsed Ewing sarcoma. Pediatr Blood Cancer. 2014;61(8):1382–1386. doi:10.1002/pbc.25042
80. Barker LM, Pendergrass TW, Sanders JE, Hawkins DS. Survival after recurrence of Ewing’s sarcoma family of tumors. J Clin Oncol. 2005;23(19):4354–4362. doi:10.1200/JCO.2005.05.105
81. McTiernan A, Driver D, Michelagnoli MP, Kilby AM, Whelan JS. High dose chemotherapy with bone marrow or peripheral stem cell rescue is an effective treatment option for patients with relapsed or progressive Ewing’s sarcoma family of tumours. Ann Oncol. 2006;17(8):1301–1305. doi:10.1093/annonc/mdl108
82. Whelan J, Le Deley MC, Dirksen U, et al. High-dose chemotherapy and blood autologous stem-cell rescue compared with standard chemotherapy in localized high-risk Ewing sarcoma: results of Euro-E.W.I.N.G.99 and Ewing-2008. J Clin Oncol. 2018;36(31):3110–3119. doi:10.1200/JCO.2018.78.251.
83. Gorlick R, Janeway KA, Adamson PC. Dose intensification improves the outcome of Ewing sarcoma. J Clin Oncol. 2018;38(31):3072–3073. doi:10.1200/JCO.2018.79.3489.
84. Jiang Y, Subbiah V, Janku F, et al. Novel secondary somatic mutations in Ewing’s sarcoma and desmoplastic small round cell tumors. PLoS One. 2014;9(8):e93676. doi:10.1371/journal.pone.0093676
Source: OncoTargets and Therapy.
Originally published March 27, 2019.