Scolaris Content Display Scolaris Content Display

Quimioterapia de dosis alta seguida de trasplante autólogo de células hematopoyéticas para niños, adolescentes y adultos jóvenes con sarcoma de Ewing primario metastásico

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Dirksen 2019 {published data only}10.1200/JCO.19.00915

Combination Chemotherapy With or Without Peripheral Stem Cell Transplantation, Radiation Therapy, and/or Surgery in Treating Patients With Ewing's Sarcoma. ClinicalTrials.gov identifier: NCT00020566. CENTRAL
Dirksen U, Brennan B, Le Deley MC, Cozic N, van den Berg H, Bhadri V, et al, Euro-EWING 99 and Ewing 2008 Investigators. High-dose chemotherapy compared with standard chemotherapy and lung radiation in Ewing sarcoma with pulmonary metastases: results of the European Ewing Tumour Working Initiative of National Groups, 99 Trial, and EWING 2008. Journal of Clinical Oncology 2019;37(34):3192-202. CENTRAL [DOI: 10.1200/JCO.19.00915]
Study in Localized and Disseminated Ewing Sarcoma (EWING2008). ClinicalTrials.gov identifier: NCT00987636. CENTRAL

Referencias de los estudios excluidos de esta revisión

Al‐Faris 2007 {published data only}10.1002/pbc.21140

Al-Faris N, Al Harbi T, Goia C, Pappo A, Doyle J,  Gassas A. Does consolidation with autologous stem cell transplantation improve the outcome of children with metastatic or relapsed Ewing sarcoma? Pediatric Blood & Cancer 2007;49(2):190-5. CENTRAL

Avramova 2006 {published data only}

Avramova B,  Jordanova M, Michailov G, Konstantinov D, Christosova I, Bobev Dr. Myeloablative chemotherapy with autologous peripheral blood stem cell transplantation in patients with poor-prognosis solid tumors – Bulgarian experience. Journal of B.U.ON. 2006;11(4):433-8. CENTRAL

Avramova 2011 {published data only}

Avramova B, Jordanova M,  Konstantinov D, Hristozova I, Shtarbanov I, Bobev D. Comparison of the treatment results after conventional and myeloablative chemotherapy in patients with poor prognosis Ewing's sarcoma family tumors - single center experience. Journal of B.U.ON. 2011;16(3):551-6. CENTRAL

Burdach 1991 {published data only}

Burdach S, Peters C, Paulussen M, Nurnberger W, Wurm R,  Wernet P, et al. Improved relapse free survival in patients with poor prognosis Ewing's sarcoma after consolidation with hyperfractionated total body irradiation and fractionated high dose melphalan followed by high dose etoposide and hematopoietic rescue. Bone Marrow Transplantation 1991;7(Suppl 2):95. CENTRAL

Burdach 1993 {published data only}10.1200/JCO.1993.11.8.1482

Burdach S, Jurgens H, Peters C, Nurnberger W, Mauz-Korholz C, Korholz D, et al. Myeloablative radiochemotherapy and hematopoietic stem-cell rescue in poor-prognosis Ewing's sarcoma. Journal of Clinical Oncology 1993;11(8):1482-8. CENTRAL [DOI: 10.1200/JCO.1993.11.8.1482]

Clara 2012 {published data only}10.1038/bmt.2012.37

Clara A,  Moita F, Ferreira I, Teixeira G, Miranda N, Leal Da Costa F, et al. Autologous stem cell transplantation for high-risk Ewing's sarcoma: a single-centre review. Bone Marrow Transplantation 2012;47:S425. CENTRAL [DOI: 10.1038/bmt.2012.37]

Cristofani 2013 {published data only}10.1111/petr.12037

Cristofani LM, Fernandes JF, Almeida MTA, Cornacchioni ALB, Azambuja AP, Gomes A, et al. Megachemotherapy with hematopoietic stem cell rescue for the treatment of children with advanced solid tumors. Pediatric Transplantation 2013;17(2):192. CENTRAL [DOI: 10.1111/petr.12037]

Diaz 2010 {published data only}10.3109/08880011003639994

Diaz MA, Lassaletta A, Perez A, Sevilla J, Madero L, Gonzalez-Vicent M. High-dose busulfan and melphalan as conditioning regimen for autologous peripheral blood progenitor cell transplantation in high-risk Ewing sarcoma patients: A long-term follow-up single-center study. Pediatric Hematology and Oncology 2010;27(4):272-82. CENTRAL [DOI: 10.3109/08880011003639994]

Dirksen 2016 {published data only}

Dirksen U, Le Deley MC, Brennan B, Judson IR, Bernstein ML, Gorlick RG, et al. Efficacy of busulfan-melphalan high dose chemotherapy consolidation (BuMel) compared to conventional chemotherapy combined with lung irradiation in Ewing sarcoma (ES) with primary lung metastases: Results of EURO-EWING 99-R2pulm randomized trial (EE99R2pul). Journal of Clinical Oncology 2016;34(15_Suppl):11001. CENTRAL [DOI: 10.1200/JCO.2016.34]

Drabko 2005 {published data only}10.1111/j.1399-3046.2005.00359.x

Drabko K, Zawitkowska-Klaczynska J, Wojcik B, Choma M, Zaucha-Prazmo A, Kowalczyk J, et al. Megachemotherapy followed by autologous stem cell transplantation in children with Ewing's sarcoma. Pediatric Transplantation 2005;9(5):618-21. CENTRAL [DOI: 10.1111/j.1399-3046.2005.00359.x]

Drabko 2006 {published data only}

Drabko K, Choma M, Zaucha-Prazmo A, Wojcik B, Gorczynska E, Kalwak K, et al. Megachemotherapy and autologous hematopoietic stem cell transplantation in children with solid tumours excluding neuroblastoma – experience of Polish paediatric centres. Medycyna wieku rozwojowego 2006;10(3):785-92. CENTRAL

Drabko 2009 {published data only}10.1002/pbc.22234

Drabko K, Zaucha-Prazmo A, Choma M, Wojcik B, Dyla A, Kalwak K, et al. Eficacy of megachemotherapy and autologous stem cell transplantation in patients with Ewing sarcoma highrisk group. Pediatric Blood & Cancer November 2009;53(5):842-3. CENTRAL [DOI: 10.1002/pbc.22234]

Drabko 2012 {published data only}10.1038/bmt.2012.78

Drabko K, Raciborska A, Bilska K, Styczynski J, Ussowicz M, Choma M, et al. Consolidation of first-line therapy with busulphan and melphalan, and autologous stem cell rescue in children with Ewing's sarcoma. Bone Marrow Transplantation 2012;47(12):1530-4. CENTRAL [DOI: 10.1038/bmt.2012.78]

Drabko 2015 {published data only}10.1038/bmt.2015.29

Drabko KA, Raciborska A, Bilska K, Ussowicz M, Styczynski J, Zaucha-Prazmo A, et al. Busulfan and treosulfan have a comparable efficacy in patients with Ewing sarcoma – retrospective analysis of Polish pediatric centers. Bone Marrow Transplantation 2015;50:S308. CENTRAL [DOI: 10.1038/bmt.2015.29]

Elhasid 2012 {published data only}10.1038/bmt.2012.37

Elhasid R, Bitan M, Cohen-Neuman N, Porat I, Furer R, Levin D, et al. Autologous stem cell transplantation in Ewing's sarcoma: the experience of Rambam Medical Centre and Tel Aviv Medical Centre. Bone Marrow Transplantation 2012;47:S425-6. CENTRAL [DOI: 10.1038/bmt.2012.37]

Engelhardt 2007 {published data only}10.1007/s00432-006-0137-1

Engelhardt M,  Zeiser R, Ihorst G, Finke J, Muller CI. High-dose chemotherapy and autologous peripheral blood stem cell transplantation in adult patients with high-risk or advanced Ewing and soft tissue sarcoma. Journal of Cancer Research and Clinical Oncology 2007;133(1):1-11. CENTRAL [DOI: 10.1007/s00432-006-0137-1]

Escobar 2010 {published data only}10.1002/pbc.22779

Escobar NF, Saa G, Trentadue J, Drelichman G, Jaimovich G, Guiterrez M, et al. Autologous hematopoietic stem cell transplantation for solid tumors. One group experience. Pediatric Blood & Cancer 2010;55(5):930-1. CENTRAL [DOI: 10.1002/pbc.22779]

Fraser 2006 {published data only}10.1038/sj.bmt.1705224

Fraser CJ, Weigel BJ, Perentesis JP, Dusenbery KE, DeFor TE, Baker KS, et al. Autologous stem cell transplantation for high-risk Ewing's sarcoma and other pediatric solid tumors. Bone Marrow Transplantation 2006;37(2):175-81. CENTRAL [DOI: 10.1038/sj.bmt.1705224]

Frohlich 1999 {published data only}10.1055/s-2008-1043801

Frohlich B, Ahrens S, Burdach S, Klingebiel T, Ladenstein R, Paulussen M, et al. High-dosage chemotherapy in primary metastasized and relapsed Ewing's sarcoma. (EI)CESS [Hochdosistherapie bei primär metastasiertem und rezidiviertem Ewing-sarkom. (EI)CESS]. Klinische Padiatrie 1999;211(4):284-90. CENTRAL [DOI: 10.1055/s-2008-1043801]

Gardner 2008 {published data only}10.1038/bmt.2008.2

Gardner SL, Carreras J, Boudreau C,  Camitta BM, Adams RH, Chen AR, et al. Myeloablative therapy with autologous stem cell rescue for patients with Ewing sarcoma. Bone Marrow Transplantation 2008;41(10):867-72. CENTRAL [DOI: 10.1038/bmt.2008.2]

Hotte 2004 {published data only}10.1080/13577140410001710521

Hotte SJ, Smith AM, Bramwell VHC, Howson-Jan K. High-dose chemotherapy followed by peripheral and/or bone marrow stem cell transplant in patients with advanced sarcoma: experience of a Canadian centre. Sarcoma 2004;8(2-3):63-9. CENTRAL [DOI: 10.1080/13577140410001710521]

Ivanova 2012 {published data only}10.1007/s00520-012-1479-7

Ivanova N, Shvarova A, Dolgopolov I. The treatment of children with primary metastatic Ewing sarcoma family tumors. Supportive Care in Cancer 2012;20:S87. CENTRAL [DOI: 10.1007/s00520-012-1479-7]

Jodele 2010 {published data only}10.1016/j.bbmt.2009.12.108

Jodele S, Davies SM, Wagner LM, Mehta PA, Bleesing JJ, Filipovich AH, et al. Outcomes with high dose chemotherapy and autologous stem cell transplantation (ASCT) in comparison to conventional chemotherapy in pediatric patients with high risk Ewing's family tumors. Biology of Blood and Marrow Transplantation 2010;16(2 Suppl 2):S186. CENTRAL [DOI: 10.1016/j.bbmt.2009.12.108]

Jurgens 2012 {published data only}10.1007/s00432-011-1144-4

Jurgens H, Dirksen U, Jabar S, Ehlert K, Boos J, Bader P, et al. Retrospective analysis of treatment-related toxicities and outcome in high-risk Ewing sarcoma patients receiving treosulfan or busulfan-based high-dose chemotherapy with autologous stem cell transplantation. Journal of Cancer Research and Clinical Oncology 2012;138:9. CENTRAL [DOI: 10.1007/s00432-011-1144-4]

Kabickova 2003 {published data only}

Kabickova E. High-dose chemotherapy with autologous hematopoietic stem cell transplantation for pediatric solid tumors. Klinicka Onkologie 2003;16(Suppl 1):119-21. CENTRAL

Kasper 2004 {published data only}10.1038/sj.bmt.1704520

Kasper B, Lehnert T, Bernd L, Mechtersheimer G, Goldschmidt H, Ho AD, et al. High-dose chemotherapy with autologous peripheral blood stem cell transplantation for bone and soft-tissue sarcomas. Bone Marrow Transplantation 2004;34(1):37-41. CENTRAL [DOI: 10.1038/sj.bmt.1704520]

Kasper 2007 {published data only}10.1159/000120629

Kasper B, Dietrich S, Mechtersheimer G, Ho AD, Egerer G. Large institutional experience with dose-intensive chemotherapy and stem cell support in the management of sarcoma patients. Oncology 2007;73(1-2):58-64. CENTRAL [DOI: 10.1159/000120629]

Kazantsev 2009 {published data only}10.3205/ctt-2009-No5-abstract08

Kazantsev IV, Youhta TV, Morozova EV, Safonova SA, Punanov YA, Zubarovskaya LS, et al. High-dose chemotherapy and autologous stem cell transplantation in children with Ewing's sarcoma/PNET. Cellular Therapy and Transplantation 2009;2(5):70-1. CENTRAL [DOI: 10.3205/ctt-2009-No5-abstract08]

Kazantsev 2011 {published data only}10.1038/bmt.2011.48

Kazantsev I, Youhta T, Zinchenko A, Gevorgyan A, Morozova E,  Safonova S, et al. High-dose chemotherapy and autologous stem cell transplantation in paediatric Ewing's sarcoma/PNET. Bone Marrow Transplantation 2011;46:S384. CENTRAL [DOI: 10.1038/bmt.2011.48]

Kazantsev 2016 {published data only}10.1038/bmt.2016.46

Kazantsev I, Youkhta T, Gevorgian A, Kozlov A, Sergei S, Tolkunova P, et al. High-dose chemotherapy with auto-or allo-HSCT in high-risk Ewing sarcoma family tumors, a single center experience. Bone Marrow Transplantation 2016;51:S58. CENTRAL [DOI: 10.1038/bmt.2016.46]

Ladenstein 1995 {published data only}

Ladenstein R, Gadner H, Hartmann O, Pico J, Biron P, Thierry P. European experience with megatherapy and ABMT in poor prognosis solid tumors (Ewing's sarcoma, germ cell tumors and brain tumors). Wiener Medizinische Wochenschrift 1995;145(2-3):55-7. CENTRAL

Ladenstein 2010 {published data only}10.1200/JCO.2009.22.9864

Ladenstein R, Potschger U, Le Deley MC, Whelan J, Paulussen M, Oberlin O, et al. Primary disseminated multifocal Ewing sarcoma: results of the Euro-EWING 99 trial. Journal of Clinical Oncology 2010;28(20):3284-91. CENTRAL [DOI: 10.1200/JCO.2009.22.9864]

Ladenstein 2014 {published data only}10.1002/pbc.25314

Ladenstein R, Valteau-Couanet D, Glogova E, Juergens H, Burdach S, Michon J, et al. The role of megatherapy (MGT) and stem cell transplantation (SCT) in high risk Ewing tumors (ET): more than 30 years of EBMT activity. Pediatric Blood & Cancer 2014;61:S176. CENTRAL [DOI: 10.1002/pbc.25314]

Lopez 2011 {published data only}10.1016/j.rpor.2011.04.002

Lopez JL, Perez C, Marquez C, Cabrera P, Perez JM, Ramirez GL, et al. Myeloablative therapy against high risk Ewing's sarcoma: a single institution experience and literature review. Reports of Practical Oncology and Radiotherapy 2011;16(5):163-9. CENTRAL [DOI: 10.1016/j.rpor.2011.04.002]

Luksch 2012 {published data only}10.1093/annonc/mds117

Luksch R, Tienghi A, Sundby Hall K, Fagioli F, Picci P, Barbieri E, et al. Primary metastatic Ewing's family tumors: results of the Italian Sarcoma Group and Scandinavian Sarcoma Group ISG/SSG IV Study including myeloablative chemotherapy and total-lung irradiation. Annals of Oncology 2012;23(11):2970-6. CENTRAL [DOI: 10.1093/annonc/mds117]

Madero 1998 {published data only}10.1038/sj.bmt.1701189

Madero L, Munoz A, Sanchez de Toledo J, Diaz MA, Maldonado MS, Ortega JJ, et al. Megatherapy in children with high-risk Ewing's sarcoma in first complete remission. Bone Marrow Transplantation 1998;21(8):795-9. CENTRAL [DOI: 10.1038/sj.bmt.1701189]

MarcusJr 1988 {published data only}

Marcus RB Jr, Graham-Pole JR, Springfield DS, Fort JA, Gross S, Mendenhall NP, et al. High-risk Ewing's sarcoma: end-intensification using autologous bone marrow transplantation. International Journal of Radiation Oncology, Biology, Physics 1988;15(1):53-9. CENTRAL

Meyers 2001 {published data only}10.1200/JCO.2001.19.11.2812

Meyers PA, Krailo MD, Ladanyi M, Chan K, Sailer SL, Dickman PS, et al. High-dose melphalan, etoposide, total-body irradiation, and autologous stem-cell reconstitution as consolidation therapy for high-risk Ewing's sarcoma does not improve prognosis. Journal of Clinical Oncology 2001;19(11):2812-20. CENTRAL [DOI: 10.1200/JCO.2001.19.11.2812]

Oberlin 2006 {published data only}10.1200/JCO.2006.05.7059

Oberlin O, Rey A, Desfachelles AS, Philip T, Plantaz D, Schmitt C, et al. Impact of high-dose busulfan plus melphalan as consolidation in metastatic Ewing tumors: a study by the Societe Francaise des Cancers de l'Enfant. Journal of Clinical Oncology 2006;24(24):3997-4002. CENTRAL [DOI: 10.1200/JCO.2006.05.7059]

Pape 1999 {published data only}10.1007/s000660050058

Pape H, Laws HJ, Burdach S, Van Kaik B, Glag M, Gripp S, et al. Radiotherapy and high-dose chemotherapy in advanced Ewing's tumors. Strahlentherapie und Onkologie 1999;175(10):484-7. CENTRAL [DOI: 10.1007/s000660050058]

Paulussen 1998 {published data only}10.1023/A:1008208511815

Paulussen M, Ahrens S, Burdach S, Craft A, Dockhorn-Dworniczak B, Dunst J, et al. Primary metastatic (stage IV) Ewing tumor: survival analysis of 171 patients from the EICESS studies. Annals of Oncology 1998;9(3):275-81. CENTRAL [DOI: 10.1023/A:1008208511815]

Pawlowska 2012 {published data only}10.1016/j.bbmt.2011.12.135

Pawlowska AB, Wolfson JA, Cheng J, Sorrell A, Sato J, Anderson C, et al. High dose chemotherapy (HDT) with busulfan, melphalan and topotecan followed by autologous hematopoietic stem cell transplantation (ASCT) in pediatric patients (PTS) with high risk solid tumors. Biology of Blood and Marrow Transplantation 2012;2:S249-50. CENTRAL [DOI: 10.1016/j.bbmt.2011.12.135]

Perentesis 1999 {published data only}

Perentesis JP, Katsanis E, DeFor TE, Neglia JP, Ramsay NKC. Autologous stem cell transplantation for high-risk pediatric solid tumors. Bone Marrow Transplantation 1999;24(6):609-15. CENTRAL

Petrovitch 2009 {published data only}

Petrovitch S, Zhavrid E, Aleinikova O, Kiselev L, Strongin YU, Baranau Y, et al. Experience in high-dose chemotherapy with peripheral stem cell rescue and biotherapy for young adults with high-risk Ewing/PNET sarcoma. European Journal of Cancer 2009;7(2-3):598. CENTRAL

Pimenov 2009 {published data only}10.3205/ctt-2009-No5-abstract03

Pimenov RI, Dolgopolov IS, Boyarshinov VK, Subbotina NS, Visochin IV, Siegiel S, et al. Treatment of patients with high-risk axial and pelvic Ewing's sarcoma (ES). A single institution experience. Cellular Therapy and Transplantation 2009;2(5):94. CENTRAL [DOI: 10.3205/ctt-2009-No5-abstract03]

Pimenov 2013 {published data only}10.1002/pbc.24509

Pimenov R, Dolgopolov I, Subbotina N, Boyarshinov V, Mentkevich G. Treatment of patients with high-risk axial and pelvic Ewing's sarcoma (ES). A single institution experience. Pediatric Blood & Cancer 2013;60:S103. CENTRAL [DOI: 10.1002/pbc.24509]

Ranft 2011 {published data only}10.1038/bmt.2011.48

Ranft A, Jabar S, Dirksen U, Ehlert K, Boos J, Bader P, et al. Retrospective analysis of treatment-related toxicities and outcome in high-risk Ewing sarcoma patients receiving PO or IV busulfan-based high-dose chemotherapy with autologous stem cell transplantation. Bone Marrow Transplantation 2011;46:S381. CENTRAL [DOI: 10.1038/bmt.2011.48]

Rose 2009 {published data only}10.1002/pbc.22234

Rose A, Zubizarreta P, Innocenti S, Raslawski E, De Davila MTG, Marti JL, et al. Metastatic Ewing/PNET sarcoma of bone: a 12-year experience in a single institution following the guidelines of Societe Francaise d'Oncologie Pediatrique EW-93 trial. Pediatric Blood & Cancer 2009;53(5):797. CENTRAL [DOI: 10.1002/pbc.22234]

Seitz 2019 {published data only}10.1002/pbc.27884

Seitz G, Urla C, Sparber-Sauer M, Schuck A, Vokuhl C, Blank B, et al. Treatment and outcome of patients with thoracic tumors of the Ewing sarcoma family: a report from the Cooperative Weichteilsarkom Studiengruppe CWS-81, -86, -91, -96, and -2002P trials. Pediatric Blood & Cancer 2019;66(Suppl 3):e27884. CENTRAL [DOI: 10.1002/pbc.27884]

Seo 2013 {published data only}10.3345/kjp.2013.56.9.401

Seo J, Kim DH, Lim JS, Koh JS, Yoo JY, Kong CB, et al. High-dose chemotherapy and autologous peripheral blood stem cell transplantation in the treatment of children and adolescents with Ewing sarcoma family of tumors. Korean Journal of Pediatrics 2013;56(9):401-6. CENTRAL [DOI: 10.3345/kjp.2013.56.9.401]

Stradella 2011 {published data only}

Stradella A, Lopez-Pousa A, Quintana MJ, Murata P,  Ortin M, Gallego O, et al. High-dose chemotherapy plus autologous stem cell transplantation (HDCT/SCT) in patients with sarcoma: a single institution experience. Journal of Clinical Oncology 2011;29(15_Suppl):10042. CENTRAL [DOI: 10.1200/jco.2011.29]

Suzuki 2015 {published data only}10.1093/annonc/mdv472.156

Suzuki R, Takahashi Y, Inoue M, Kanamori H, Hashii Y, Sakamaki H, et al. Thiotepa for autologous hematopoietic stem cell transplantation for solid tumors. Annals of Oncology 2015;26:vii140-1. CENTRAL [DOI: 10.1093/annonc/mdv472.156]

Tanaka 2002 {published data only}10.1007/s007760200083

Tanaka K, Matsunobu T, Sakamoto A, Matsuda S, Iwamoto Y. High-dose chemotherapy and autologous peripheral blood stem-cell transfusion after conventional chemotherapy for patients with high-risk Ewing's tumors. Journal of Orthopaedic Science 2002;7(4):477-82. CENTRAL [DOI: 10.1007/s007760200083]

Wessalowski 1988 {published data only}10.1055/s-2008-1033717

Wessalowski R, Jurgens H, Bodenstein H, Brandeis W, Gutjahr P, Havers W, et al. Results of treatment of primary metastatic Ewing sarcoma. A retrospective analysis of 48 patients. Klinische Padiatrie 1988;200(3):253-60. CENTRAL [DOI: 10.1055/s-2008-1033717]

Whelan 2018 {published data only}10.1200/JCO.2018.78.2516

Whelan J, Le Deley MC, Dirksen U, Le Teuff G, Brennan B, Gaspar N, et al, Euro-EWING99 and EWING-2008 Investigators. 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. Journal of Clinical Oncology 2018;36:3110-9. CENTRAL [PMID: 10.1200/JCO.2018.78.2516]

Referencias de los estudios en espera de evaluación

Dirksen 2020 {published data only}10.1200/JCO.2020.38.15_suppl.11501

Dirksen U, Bhadri V, Brichard B, Butterfass-Bahloul T, Cyprova S, Faldum A, et al, on behalf of the Cooperative Ewing Sarcoma Study Group Ewing 2008, Sponsor UKM. Efficacy of add-on treosulfan and melphalan high-dose therapy in patients with high-risk metastatic Ewing sarcoma: report from the International Ewing 2008R3 trial. Journal of Clinical Oncology 2020;38(15):11501. CENTRAL [DOI: 10.1200/JCO.2020]

Bacci 2000

Bacci G, Ferrari S, Bertoni F, Rimondini S, Longhi A, Bacchini P, et al. Prognostic factors in non metastatic Ewing's sarcoma of bone treated with adjuvant chemotherapy: analysis of 359 patients at the Istituto Ortopedico Rizzoli. Journal Clinical Oncology 2000;18(1):4-11.

Burdach 2000

Burdach S, van Kaick B, Laws HJ, Ahrens S, Haase R, Körholz D, et al, the Stem-Cell Transplant Programs at Düsseldorf University Medical Center, Germany and St Anna Kinderspital, Vienna, Austria. Allogeneic and autologous stem-cell transplantation in advanced Ewing tumours. An update after long-term follow-up from two centers of the European Intergroup study EICESS. Annals of Oncology 2000;11(11):1451-62.

Cangir 1990

Cangir A, Vietti TJ, Gehan EA, Burgert EO Jr, Thomas P, Tefft M, et al. Ewing's sarcoma metastatic at diagnosis. Results and comparisons of two intergroup Ewing's sarcoma studies. Cancer 1990;66(5):887-93.

Carli 2004

Carli M, Colombatti R, Oberlin O, Bisogno G, Treuner J, Koscielniak E, et al. European intergroup studies (MMT4-89 and MMT4-91) on childhood metastatic rhabdomyosarcoma: final results and analysis of prognostic factors. Journal of Clinical Oncology 2004;22(23):4787-94.

Claviez 2008

Claviez A, Sureda A, Schmitz N. Haematopoietic SCT for children and adolescents with relapsed and refractory Hodgkin's lymphoma. Bone Marrow Transplantation 2008;42(Suppl 2):S16-24.

Cotterill 2000

Cotterill SJ, Ahrens S, Paulussen M, Jürgens HF, Voûte PA, Gadner H, et al. Prognostic factors in Ewing's tumor of bone: analysis of 975 patients from the European Intergroup Cooperative Ewing's Sarcoma Study Group. Journal Clinical Oncology 2000;18(17):3108-14.

Delattre 1994

Delattre O, Zucman J, Melot T. The Ewing family of tumors – a subgroup of small-round-cell tumors defined by specific chimeric transcripts. New England Journal of Medicine 1994;331(5):294-9.

Esiashvili 2008

Esiashvili N, Goodman M, Marcus RB Jr. Changes in incidence and survival of Ewing sarcoma patients over the past 3 decades: surveillance epidemiology and end results data. Journal of Pediatric Hematology/Oncology 2008;30(6):425-30.

Gaspar 2015

Gaspar N, Hawkins DS, Dirksen U, Lewis IJ, Ferrari S, Le Deley MC, et al. Ewing Sarcoma: current management and future approaches through collaboration. Journal of Clinical Oncology Sep 2015;33(27):3036-46. [DOI: 10.1200/JCO.2014.59.5256]

Gorlick 2013

Gorlick R, Janeway K, Lessnick S, Randall RL, Marina N, COG Bone Tumor Committee. Children's Oncology Group's 2013 blueprint for research: bone tumors. Pediatric Blood Cancer 2013;60(6):1009-15.

GRADE Handbook

Schünemann H, Brożek J, Guyatt G, Oxman A, editor(s). GRADE handbook for grading quality of evidence and strength of recommendation. Version 3.6 (updated October 2011). The GRADE Working Group. 2011.

GRADEpro GDT [Computer program]

McMaster University (developed by Evidence Prime)GRADEpro GDT. Version accessed prior to 15 October 2020. Hamilton (ON): McMaster University (developed by Evidence Prime). Available from gradepro.org.

Gurney 1999

Gurney JG, Swensen AR, Bulterys M. Malignant bone tumours. In: Ries LAG, Smith MA, Gurney JG, Linet M, Tamra T, Young JL, et al, editors(s). Cancer Incidence and Survival Among Children and Adolescents: United States SEER Program 1975-1999. Bethesda, MD: National Cancer Institute SEER Program, National Institutes of Health, 1999:99-110.

Haeusler 2010

Haeusler J, Ranft A, Boelling T, Gosheger G, Braun-Munzinger G, Vieth V, et al. The value of local treatment in patients with primary, disseminated, multifocal Ewing sarcoma. Cancer 2010;116:443-50.

Higgins 2011

Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from training.cochrane.org/handbook/archive/v5.1/.

Jenkin 2001

Jenkin RD, Al-Fawaz I, Al-Shabanah MO, Allam A, Ayas M, Memon M, et al. Metastatic Ewing sarcoma/PNET of bone at diagnosis: prognostic factors – a report from Saudi Arabia. Medical and Pediatric Oncology 2001;37(4):383-9.

Kauer 2009

Kauer M, Ban J, Kofler R, Walker B, Davis S, Meltzer P, et al. A molecular function map of Ewing's sarcoma. PloS One 2009;4(4):e5415.

Kushner 2001

Kushner BH, Meyers PA. How effective is dose-intensive/myeloablative therapy against Ewing's sarcoma/primitive neuroectodermal tumor metastatic to bone or bone marrow? The Memorial Sloan-Kettering experience and a literature review. Clinical Oncology 2001;19(3):870-80.

Lee 2011

Lee JA, Kim DH, Cho J, Lim JS, Koh JS, Yoo JY, et al. Treatment outcome of Korean patients with localized Ewing sarcoma family of tumors: a single institution experience. Japanese Journal of Clinical Oncology 2011;41(6):776-82.

Matthay 1999

Matthay KK. Intensification of therapy using hematopoietic stem-cell support for high-risk neuroblastoma. Pediatric Transplantation 1999;3(Suppl 1):72-7.

Module CCG 2010

Kremer LCM, van Dalen EC, Moher D, Caron HN. Cochrane Childhood Cancer Group. About The Cochrane Collaboration (Cochrane Review Groups (CRGs))2010;(12).

Ordóñez 2009

Ordóñez JL, Osuna D, Herrero D, de Álava E, Madoz-Gúrpide J. Advances in Ewing's sarcoma research: where are we now and what lies ahead? Cancer Research 2009;69(18):7140-50.

Potratz 2012

Potratz J, Dirksen U, Jürgens H, Craft A. Ewing sarcoma: clinical state-of-the-art. Pediatric Hematology and Oncology 2012;29:1-11.

Review Manager 2020 [Computer program]

The Cochrane CollaborationReview Manager 5 (RevMan 5). Version 5.4. The Cochrane Collaboration, 2020.

Rosenthal 2008

Rosenthal J, Bolotin E, Shakhnovits M, Pawlowska A, Falk P. High-dose therapy with hematopoietic stem cell rescue in patients with poor prognosis Ewing family tumors. Bone Marrow Transplantation 2008;42(5):311-8.

Spreafico 2008

Spreafico F, Bisogno G, Collini P, Jenkner A, Gandola L, D'Angelo P. Treatment of high-risk relapsed Wilms tumor with dose-intensive chemotherapy, marrow-ablative chemotherapy, and autologous hematopoietic stem cell support: experience by the Italian Association of Pediatric Hematology and Oncology. Pediatric Blood Cancer 2008;51(1):23-8.

Teicher 2011

Teicher BA, Bagley RG, Rouleau C, Kruger A, Ren Y, Kurtzberg L. Characteristics of human Ewing/PNET sarcoma models. Annals of Saudi Medicine 2011;31(2):174-82.

Van den Berg 2008

Van den Berg H, Kroon HM, Slaar A, Hogendoorn P. Incidence of biopsy-proven bone tumors in children. A report based on the Dutch pathology registration "PALGA". Journal of Pediatric Orthopaedics 2008;28(1):29-35.

Referencias de otras versiones publicadas de esta revisión

Haveman 2014

Haveman LM, Breunis WB, van Dalen EC, Kremer LCM, Dirksen U, Jürgens H, et al. High-dose chemotherapy followed by autologous haematopoietic cell transplantation for children, adolescents and young adults with metastatic Ewing sarcoma. Cochrane Database of Systematic Reviews 2014, Issue 12. Art. No: CD011405. [DOI: 10.1002/14651858.CD011405]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Dirksen 2019

Study characteristics

Methods

The R2Pulm trial was an international, randomised, superiority trial comparing two consolidation regimens in a two‐parallel‐group design. The R2Pulm randomised trial was part of the Euro‐E.W.I.N.G. 99 study (ClinicalTrials.gov identifier: NCT00020566), recruiting people with Ewing sarcoma at diagnosis. From May 2010, people were also recruited in the same R2Pulm randomised trial conducted through the EWING 2008 study (ClinicalTrials.gov identifier: NCT00987636)

Enrolment took place in 144 centres from 14 countries between December 2015 and February 2000.

Randomisation was completed after four to six combed vincristine, ifosfamide, doxorubicin, and etoposide (VIDE) courses. It was balanced and stratified according to co‐operative group, sex, age (younger than 25 years), and local treatment (surgery after induction chemotherapy with or without postoperative radiotherapy versus initial surgery versus combined surgery after induction chemotherapy followed by radiotherapy versus radiotherapy only).

Central randomisation software was used in all data centres, ensuring the concealment of the next participant allocation. The Gesellschaft fur Paediatrische Onkologie und Haematologie data centre used permuted blocks of four. In the European Organisation for Research and Treatment of Cancer, Children’s Cancer and Leukaemia Group, and French data centres, randomisation was also balanced by the treating centre using dynamic allocation of treatment (minimisation with a random factor set at 0.8).

Participants

Eligible participants were younger than 50 years of age. Therefore, we only included data if described separately for our included population (younger than 30 years of age). In descriptive data and analyses, this study classified the following age categories: < 12 years; 12 to 18 years; 18 to 25 years, and > 25 years. 

There were 267 participants younger than 25 years of age, and diagnosed with Ewing sarcoma with pulmonary or pleural metastases, without other distant metastases included in this study.

Age:

Not reported specifically for eligible patients

  • the whole study group: median age 14.2 years (range, 1.0 to 47.8 years)

  • intervention group: median 13.9 years (range 1.7 to 47.8 years)

  • control group: median 14.6 years (range 1.0 to 44.1 years)

Gender:

Not reported specifically for eligible participants

  • the whole study group: 59% male

  • intervention group: 60% male

  • control group: 57% male

Known risk factors for people with metastatic disease:

  • Tumour volume: not reported for eligible participants

    • the whole study group: 60% ≥ 200 mL

    • equal in both treatment arms: 60% ≥ 200 mL

  • Sites of metastases (lungs, bone, bone marrow, or combination, were not reported for eligible participants

    • the whole study group: 12% single pulmonary nodules and 88% multiple pulmonary nodules;

    • intervention group 89% multiple nodes

    • control group 86% multiple nodes

    • there were no bone or bone marrow metastases

Interventions

For all participants, induction chemotherapy consisted of six VIDE chemotherapy courses and 1 combined vincristine, actinomycin D, and Ifosfamide (VAI) course.

After induction, the allocated consolidation treatment was:

  • control group: seven courses of VAI followed by WLI for the control arm (N = 134)

  • intervention group: one course of combined high‐dose busulfan and melphalan (BuMel) chemotherapy with autologous stem‐cell transplantation (N = 133)

Participants aged younger than 12 years: chemotherapy and WLI (N = 44) versus BuMel and AHCT (N = 48)

Participants aged 12 to 18 years: chemotherapy and WLI (N = 66) versus BuMel and AHCT (N = 62)

Participants aged 18 to 25 years: chemotherapy and WLI (N = 24) versus BuMel and AHCT (N = 23)

Local therapy was tailored to participant and tumour characteristics, and included complete surgical removal of the tumour wherever feasible, radiotherapy, or a combination of both.

In the BuMel (intervention) group

  • 115 participants (80%) underwent resection after chemotherapy, with or without late radiotherapy

  • 29 people received other local treatment: 4 had a resection at diagnosis, and 22 received radiotherapy alone

In the conventional chemotherapy with WLI (control) group,

  • 120 participants (84%)

  • 23 people with other local treatment: 3 underwent resection after chemotherapy and early RT, 1 underwent resection at diagnosis, and 16 received radiotherapy alone

Autologous haematopoietic stem‐cell harvesting was completed according to local practices, at the earliest, after VIDE course 2

No actually received cumulative doses of the different chemotherapeutics were described. As described in the Ewing 2008 protocol the courses consisted of: 

VIDE courses (6 for each group)

  • Vincristine: 1.5 mg/m², 6 cycles = cumulative dose of 9 mg/m²

  • Ifosfamide: 9000 mg/m², 6 cycles = cumulative dose of 54,000 mg/m²

  • Doxorubicin: 60 mg/m², 6 cycles = cumulative dose of 360 mg/m²

  • Etoposide: 450 mg/m²; 6 cycles = cumulative dose of 2700 mg/m²

VAI courses (1 for intervention group, 8 for control group)

Intervention group:

  • Vincristine 1.5 mg/m², 1 cycle = cumulative dose of 1.5 mg/m²

  • Actinomycin D: 1.5 mg/m², 1 cycle = cumulative dose of 1.5 mg/m²

  • Ifosfamide: 6000mg/m², 1 cycle = cumulative dose of 6000 mg/m²

Control group:

  • Vincristine 1.5 mg/m²,  8 cycles = cumulative dose of 12 mg/m²

  • Actinomycin D: 1.5 mg/m², 8 cycles = cumulative dose of 12 mg/m²

  • Ifosfamide: 6000mg/m², 8 cycles = cumulative dose of 48,000 mg/m²

BuMel course

Busulfan: 

  • Adults: 12.8 mg/kg

  • Children and adolescents: < 9 kg = 16 mg/kg; 9 kg ≤ 16 kg = 19.2 mg/kg; 16 kg to 23 kg = 17.6 mg/kg; 23 kg to 34 kg = 15.2 mg/kg; > 34 kg = 12.8 mg/kg

Melphalan: 140 mg/m²  

Outcomes

Event‐free survival (defined as the time from randomisation to the time of the first event assessed by the investigator, defined as progression, relapse, second malignancy, or death, whatever the cause).

Notes

No information provided on follow‐up for the eligible participants (for the whole study group median 8.1 years (range 0 to 15.5 years), similar in the intervention group versus the conventional chemotherapy with WLI group).

Overall survival and toxicity data were assessed in the study, but not separately for the eligible participants for this review. The study authors were unable to provide us with additional data.

The study was supported by FP7‐EURO EWING Consortium; Association Enfants et Sante, Societe Française de Lutte Contre les Cancers et les Leucemies de l’Enfant et de l’Adolescent, Unicancer; Cancer Research UK (CRUK/02/ 014), National Institute for Health Research, University College London Hospitals, Biomedical Research Centre; Deutsche Krebshilfe (70‐2551‐Jue3 and 108128), and Bundesministerium fur Bildung und Forschung (BMBF 01GM0869; BMBF/Era‐Net 01KT1310); National Cancer Institute, Bethesda, MD (U10CA098413, U10CA098543, U10CA180886, and U10CA180899); and The Swedish Childhood Cancer Fund.

Authors' disclosures of potential conflict of interest are described. Study authors were sponsored by pharma.

No other potential conflicts of interest were reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation software was used, ensuring random sequence generation. 

Allocation concealment (selection bias)

Low risk

Central randomisation software was used, ensuring the concealment of the next participant allocation. 

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Blinding to therapy could not be achieved because of obvious treatment differences.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information on blinding of outcome assessors of EFS was provided.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No data provided specifically for the participants eligible for this review, but out of 287 participants in the total study group, 11 in the intervention group, and 7 in the control group were lost to follow‐up. Even if all of these participants lost to follow‐up were ≤ 25 years old, the risk of attrition bias for EFS would still be low, as a maximum of 11/133 (8%) of the intervention group and 7/134 (5%) of the control group would be lost to follow‐up.

Selective reporting (reporting bias)

Low risk

Study protocol was available, and all of the study’s prespecified (primary and secondary) outcomes of interest were reported as pre‐specified. For this review, all outcomes that one would expect to be reported were defined as overall survival and toxicity. All expected outcomes were reported (although not eligible for inclusion in the review).

Other bias

High risk

Block randomisation in unblinded trials: yes

Baseline imbalance in treatment arms related to outcome (i.e. age, tumour volume): no

Important known prognostic characteristics were equally distributed: age was not reported separately for eligible participants, but the median age for the whole study group was similar in both treatment groups. Primary tumour volume was not reported separately for eligible participants, but for the whole study group; it was similar in both treatment groups. Therefore we judged this to be a low risk of bias.

Difference in length of follow‐up between treatment arms: no

No specific follow‐up was described for eligible age groups. In the whole cohort, both treatment groups had an equal follow‐up duration of 8.1 years (0 to 15.5 years). We judged this to be a low risk of bias.

Other potential biases: unclear (no information available separately for the participants eligible for this review, but in the total group, out of 543 eligible people, only 287 were enrolled in the trial for randomisation: 98 people/parents refused enrolment, 71 people were not enrolled because the physician refused, 63 people were not enrolled for miscellaneous reasons, and 24 people were not enrolled for unknown reasons). We judged this to be a high risk of bias.

WLI: whole lung irradiation; AHCT: autologous haematopoietic cell transplantation; EFS: event‐free survival

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Al‐Faris 2007

Ineligible design

Avramova 2006

Ineligible design

Avramova 2011

Ineligible design

Burdach 1991

Ineligible design

Burdach 1993

Ineligible design

Clara 2012

Ineligible design

Cristofani 2013

Ineligible design

Diaz 2010

Ineligible design

Dirksen 2016

Abstract, full study publication Dirksen 2019

Drabko 2005

Ineligible design

Drabko 2006

Ineligible design

Drabko 2009

Ineligible design

Drabko 2012

Ineligible design

Drabko 2015

Ineligible design

Elhasid 2012

Ineligible design

Engelhardt 2007

Ineligible design

Escobar 2010

Ineligible design

Fraser 2006

Ineligible design

Frohlich 1999

Ineligible design

Gardner 2008

Ineligible design

Hotte 2004

Ineligible design

Ivanova 2012

Ineligible design

Jodele 2010

Ineligible design

Jurgens 2012

Ineligible design

Kabickova 2003

Ineligible design

Kasper 2004

Ineligible design

Kasper 2007

Ineligible design

Kazantsev 2009

Ineligible design

Kazantsev 2011

Ineligible design

Kazantsev 2016

Ineligible design

Ladenstein 1995

Ineligible design

Ladenstein 2010

Ineligible design

Ladenstein 2014

Ineligible design

Lopez 2011

Ineligible design

Luksch 2012

Ineligible design

Madero 1998

Ineligible design

MarcusJr 1988

Ineligible design

Meyers 2001

Ineligible design

Oberlin 2006

Ineligible design

Pape 1999

Ineligible design

Paulussen 1998

Ineligible design

Pawlowska 2012

Ineligible design

Perentesis 1999

Ineligible design

Petrovitch 2009

Ineligible design

Pimenov 2009

Ineligible design

Pimenov 2013

Ineligible design

Ranft 2011

Ineligible design

Rose 2009

Ineligible design

Seitz 2019

Ineligible design

Seo 2013

Ineligible design

Stradella 2011

Ineligible design

Suzuki 2015

Ineligible design

Tanaka 2002

Ineligible design

Wessalowski 1988

Ineligible design

Whelan 2018

Ineligible design

Characteristics of studies awaiting classification [ordered by study ID]

Dirksen 2020

Methods

A phase III, open label, prospective, multicentre, randomised controlled clinical trial

Participants

109 people were randomised between 2009 and 2018: 55 were randomised to high‐dose TreoMel followed by autologous stem cell transplant. People had disseminated Ewing sarcoma with metastases to bone, other sites, or both, excluding people with only pleuropulmonary metastases.

Interventions

Participants received 6 cycles of VIDE induction and 8 cycles of VAC consolidation therapy. Participants were randomised to receive additional high dose chemotherapy (TreoMel) followed by autologous stem cell transplant or no further treatment (control).

Outcomes

With a median follow‐up of 3.3 years, the primary endpoint EFS was not significantly different between HDC TreoMel and the control in the adaptive design (hazard ratio (HR) 0.85, 95% confidence interval (CI) 0.55 to 1.32; intention‐to‐treat). 3‐year (3y) EFS was 20.9% (95% CI 11.5 to 37.9%) in the HDC TreoMel group, and 19.2% (95% CI 10.8 to 34.4%) in the control group. Results were similar in the per protocol collective. Subgroup analyses showed that independent of treatment, male participants had a worse outcome than female participants: 3y EFS 13.3% (95% CI 5.7 to 31.1%) vs 25.2% (95% CI 15.5 to 40.8%); P = 0.07. Participants aged < 14 yr had a better outcome when treated in the HDC TreoMel group: 3y EFS 39.3% (95% CI 20.4 to 75.8%) vs 9% (95% CI 2.4 to 34%); P = 0.016; HR 0.40 (95% CI 0.19 to 0.87). These effects were similar in the per protocol collective. Severe toxicities of hematology, gut, general condition, and infection were more pronounced in the HDC TreoMel group (P < 0.05).

Notes

This study has not been published in full text yet.

HDC: high‐dose chemotherapy; VIDE: Vincristin, Ifosfamide, Doxorubicin, Etoposide; VAC: Vincristin, Actinomycin D, Cyclophosphamide; EFS: event‐free survival

Data and analyses

Open in table viewer
Comparison 1. Survival for primary pulmonary metastatic Ewing sarcoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Event‐free survival (intention‐to‐treat analysis) Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

0.83 [0.59, 1.17]

Analysis 1.1

Comparison 1: Survival for primary pulmonary metastatic Ewing sarcoma, Outcome 1: Event‐free survival (intention‐to‐treat analysis)

Comparison 1: Survival for primary pulmonary metastatic Ewing sarcoma, Outcome 1: Event‐free survival (intention‐to‐treat analysis)

Flow diagram of selection of studiesASBMT: American Society for Blood and Marrow Transplantation; ASPHO: American Society of Pediatric Hematology/Oncology; CTOS: Connective Tissue Oncology Society; EBMT: American Society for Blood and Marrow Transplantation; EMSOS: European Musculo‐Skeletal Oncology Society; SIOP: International Society for Paediatric Oncology

Figuras y tablas -
Figure 1

Flow diagram of selection of studies

ASBMT: American Society for Blood and Marrow Transplantation; ASPHO: American Society of Pediatric Hematology/Oncology; CTOS: Connective Tissue Oncology Society; EBMT: American Society for Blood and Marrow Transplantation; EMSOS: European Musculo‐Skeletal Oncology Society; SIOP: International Society for Paediatric Oncology

original image

Figuras y tablas -
Figure 2

Forest plot of comparison: 1 Survival for primary pulmonary metastatic Ewing sarcoma, outcome: 1.1 Event‐free survival (intention‐to‐treat analysis).

Figuras y tablas -
Figure 3

Forest plot of comparison: 1 Survival for primary pulmonary metastatic Ewing sarcoma, outcome: 1.1 Event‐free survival (intention‐to‐treat analysis).

Comparison 1: Survival for primary pulmonary metastatic Ewing sarcoma, Outcome 1: Event‐free survival (intention‐to‐treat analysis)

Figuras y tablas -
Analysis 1.1

Comparison 1: Survival for primary pulmonary metastatic Ewing sarcoma, Outcome 1: Event‐free survival (intention‐to‐treat analysis)

Summary of findings 1. High‐dose chemotherapy followed by autologous haematopoietic cell transplantation compared with conventional chemotherapy plus whole lung irradiation

High‐dose chemotherapy followed by autologous haematopoietic cell transplantation compared with conventional chemotherapy plus whole lung irradiation

 

Patient or population: children, adolescents, and young adults with primary metastatic Ewing sarcoma

Settings: (paediatric) oncology departments

Intervention: high‐dose chemotherapy followed by autologous haematopoietic cell transplantation

Comparison: conventional chemotherapy plus whole lung irradiation

 

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

 

Assumed risk with CC + WLI

Corresponding risk with HDC + AHCT

 

Event‐free survival

(reported as number of participants with progression, relapse, second malignancy, or death, whatever the cause)

Follow‐up not mentioned

 

537 per 1000a

473 per 1000

(365 to 594)

HR 0.83 (0.59 to 1.17)

267
(1)

⊕⊕⊝⊝
lowb.c

 

Only pulmonary or pleural metastases, without other distant metastases, were included in this study.

Note that due to limitations of the software for this table, event‐free survival is presented as number of participants with an event.

Results in the different age groups (< 12 years, 12 to 18 years, and 18 to 25 years) were similar to the overall results described here, including the GRADE assessment.

 

Overall survival

Data for participants under the age of 30 years were not reported separately.

 

Quality‐adjusted survival

No information was provided for quality‐adjusted survival.

 

Toxicity

Data for participants under the age of 30 years were not reported separately.

 

Progression‐free survival

No information was provided for progression‐free survival.

 

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

AHCT: autologous haematopoietic cell transplantation; CC: conventional chemotherapy; CI: confidence Interval; HDC: high‐dose chemotherapy; HR: hazard ratio; WLI: whole lung irradiation

 

GRADE Working Group grades of evidence
High certainty. Further research is very unlikely to change our confidence in the estimate of effect.
Moderate certainty. Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low certainty. Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low certainty. We are very uncertain about the estimate.

 

aThe assumed risk was based on the prevalence in the control group of the included study.
bHigh risk of performance and other bias, unclear risk of detection bias (downgraded one level).
cAs this was a small study, with a total number of events fewer than 300 (the threshold rule‐of‐thumb value stated in the GRADEpro handbook (GRADE Handbook)), we downgraded one level.

Figuras y tablas -
Summary of findings 1. High‐dose chemotherapy followed by autologous haematopoietic cell transplantation compared with conventional chemotherapy plus whole lung irradiation
Comparison 1. Survival for primary pulmonary metastatic Ewing sarcoma

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Event‐free survival (intention‐to‐treat analysis) Show forest plot

1

Hazard Ratio (IV, Fixed, 95% CI)

0.83 [0.59, 1.17]

Figuras y tablas -
Comparison 1. Survival for primary pulmonary metastatic Ewing sarcoma