Scolaris Content Display Scolaris Content Display

Pokonsolidacyjna terapia kwasem retinowym chorych na agresywną postać nerwiaka zarodkowego (neuroblastoma), leczonych autologicznym przeszczepem hematopoetycznych komórek macierzystych

Contraer todo Desplegar todo

Referencias

Matthay 1999 {published data only}

Haas‐Kogan DA, Swift PS, Selch M, Haase GM, Seeger RC, Gerbing RB, et al. Impact of radiotherapy for high‐risk neuroblastoma: a Children's Cancer Group study. International Journal of Radiation Oncology Biology Physics 2003;56(1):28‐39. CENTRAL
Maris JM, Weiss MJ, Guo C, Gerbing RB, Stram DO, White PS, et al. Loss of heterozygosity at 1p36 independently predicts for disease progression but not decreased overall survival probability in neuroblastoma patients: A Children's Cancer Group study. Journal of Clinical Oncology 2000;18(9):1888‐99. CENTRAL
Matthay KK, Reynolds CP, Seeger RC, Shimada H, Adkins ES, Haas‐Kogan D, et al. Long‐term results for children with high‐risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13‐cis‐retinoic acid: a children's oncology group study. Journal of Clinical Oncology 2009;27(7):1007‐13. The publisher attached an erratum to the supplements tab of the online publication: ascopubs.org/doi/suppl/10.1200/JCO.2007.13.8925 (accessed 18 May 2017). CENTRAL
Matthay KK, Villablanca JG, Seeger RC, Stram DO, Harris RE, Ramsay NK, et al. Treatment of high‐risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13‐cis‐retinoic acid. Children's Cancer Group. New England Journal of Medicine 1999;341(16):1165‐73. CENTRAL
Park JR, Villablanca JG, London WB, Gerbing RB, Haas‐Kogan D, Adkins ES, et al. Outcome of high‐risk stage 3 neuroblastoma with myeloablative therapy and 13‐cis‐retinoic acid: a report from the Children's Oncology Group. Pediatric Blood and Cancer 2009;52(1):44‐50. CENTRAL
Schmidt ML, Lal A, Seeger RC, Maris JM, Shimada H, O'Leary M, et al. Favorable prognosis for patients 12 to 18 months of age with stage 4 nonamplified MYCN neuroblastoma: a Children's Cancer Group Study. Journal of Clinical Oncology 2005;23(27):6474‐80. CENTRAL

Adamson 1997 {published data only}

Adamson PC, Reaman G, Finklestein JZ, Feusner J, Berg SL, Blaney SM, et al. Phase I trial and pharmacokinetic study of all‐trans‐retinoic acid administered on an intermittent schedule in combination with interferon‐alpha2a in pediatric patients with refractory cancer. Journal of Clinical Oncology 1997;15(11):3330‐7. CENTRAL

Adamson 2007 {published data only}

Adamson PC, Matthay KK, O'Brien M, Reaman GH, Sato JK, Balis FM. A phase 2 trial of all‐trans‐retinoic acid in combination with interferon‐alpha2a in children with recurrent neuroblastoma or Wilms tumor: A Pediatric Oncology Branch, NCI and Children's Oncology Group Study. Pediatric Blood and Cancer 2007;49(5):661‐5. CENTRAL

Aksoylar 2013 {published data only}

Aksoylar S, Varan A, Vergin C, Hazar V, Akici F, Dagdeviren, et al. High‐dose chemotherapy and autologous stem cell transplantation in patients with high‐risk neuroblastoma : Results of Turkish pediatric oncology group‐neuroblastoma 2003 (TPOG‐NBL 2003) protocol. 55th Annual Meeting of the American Society of Hematology, ASH 2013 New Orleans, LA United States. Blood 2013;122(21):3411. CENTRAL

Anderson 2005 {published data only}

Anderson BD, Schoenfeldt M. Current phase III clinical trials investigating pediatric cancers. Oncology 2005;19:69‐74, 78. CENTRAL

Atra 1996 {published data only}

Atra A, Pinkerton R. Autologous stem cell transplantation in solid tumours of childhood. Annals of Medicine 1996;28(2):159‐64. CENTRAL

Bagatell 2014 {published data only}

Bagatell R, Fisher B, Seif AE, Huang YS, Li Y, Desai AV, et al. Evaluation of resources used during care of children with high‐risk neuroblastoma (HR NBL) via merging of cooperative group trial data and administrative data. 2014 ASCO Annual Meeting. Journal of Clinical Oncology 2014;32(5 suppl):abstr 10069. CENTRAL

Bauters 2011 {published data only}

Bauters TG, Laureys G, Van de Velde V, Benoit Y, Robays H. Practical implications for the administration of 13‐cis retinoic acid in pediatric oncology. International Journal of Clinical Pharmacotherapy 2011;33(4):597‐8. CENTRAL

Castel 2004 {published data only}

Castel V, Canete A. A comparison of current neuroblastoma chemotherapeutics. Expert Opinion on Pharmacotherapy 2004;5(1):71‐80. CENTRAL

Chan 2007 {published data only}

Chan EL, Harris RE, Emery KH, Gelfand MJ, Collins MH, Gruppo RA. Favorable histology, MYCN‐amplified 4S neonatal neuroblastoma. Pediatric Blood and Cancer 2007;48(4):479‐82. CENTRAL

Chen 2015 {published data only}

Chen SC, Murphy D, Sastry J, Shaikh MG. Predicting, monitoring, and managing hypercalcemia secondary to 13‐cis‐retinoic acid therapy in children with high‐risk neuroblastoma. Journal of Pediatric Hematology/Oncology 2015;37(6):477‐81. CENTRAL

Clarke 2003 {published data only}

Clarke E, Glaser AW, Picton SV. Pneumocystis carinii pneumonia: a late presentation following treatment for stage IV neuroblastoma. Pediatric Hematology and Oncology 2003;20(6):467‐71. CENTRAL

Cross 2009 {published data only}

Cross SF, Dalla Pozza L, Munns CF. Hypercalcemia and osteoblastic lesions induced by 13‐Cis‐retinoic acid mimicking relapsed neuroblastoma. Pediatric Blood and Cancer 2009;53(4):666‐8. CENTRAL

De Kraker 2008 {published data only}

De Kraker J, Hoefnagel KA, Verschuur AC, Van Eck B, Van Santen HM, Caron HN. Iodine‐131‐metaiodobenzylguanidine as initial induction therapy in stage 4 neuroblastoma patients over 1 year of age. European Journal of Cancer 2008;44(4):551‐6. CENTRAL

Di Bella 2009 {published data only}

Di Bella G, Colori B. Complete objective response of neuroblastoma to biological treatment. Neuroendocrinology Letters 2009;30(4):437‐49. CENTRAL

Dmitrovsky 2004 {published data only}

Dmitrovsky E. Fenretinide activates a distinct apoptotic pathway. Journal of the National Cancer Institute 2004;96(17):1264‐5. CENTRAL

Elimam 2006 {published data only}

Elimam NA, Atra AA, Fayea NY, Al‐Asaad TG, Khattab TM, Al‐Sulami GA, et al. Stage 4S neuroblastoma, a disseminated tumor with excellent outcome. Saudi Medical Journal 2006;27(11):1734‐6. CENTRAL

Finklestein 1992 {published data only}

Finklestein JZ, Krailo MD, Lenarsky C, Ladisch S, Blair GK, Reynolds CP, et al. 13‐cis‐retinoic acid (NSC 122758) in the treatment of children with metastatic neuroblastoma unresponsive to conventional chemotherapy: report from the Childrens Cancer Study Group. Medical and Pediatric Oncology 1992;20(4):307‐11. CENTRAL

Formelli 2008 {published data only}

Formelli F, Cavadini E, Luksch R, Garaventa A, Villani MG, Appierto V, et al. Pharmacokinetics of oral fenretinide in neuroblastoma patients: indications for optimal dose and dosing schedule also with respect to the active metabolite 4‐oxo‐fenretinide. Cancer Chemotherapy and Pharmacology 2008;62(4):655‐65. CENTRAL

Formelli 2010 {published data only}

Formelli F, Cavadini E, Luksch R, Garaventa A, Appierto V, Persiani S. Relationship among pharmacokinetics and pharmacodynamics of fenretinide and plasma retinol reduction in neuroblastoma patients. Cancer Chemotherapy and Pharmacology 2010;66(5):993‐8. CENTRAL

Fouladi 2010 {published data only}

Fouladi M, Park JR, Stewart CF, Gilbertson RJ, Schaiquevich P, Sun J, et al. Pediatric phase I trial and pharmacokinetic study of vorinostat: a Children's Oncology Group phase I consortium report. Journal of Clinical Oncology 2010;28(22):3623‐9. CENTRAL

French 2013 {published data only}

French S, Dubois SG, Horn B, Granger M, Hawkins R, Pass A, et al. 131I‐MIBG followed by consolidation with busulfan, melphalan and autologous stem cell transplantation for refractory neuroblastoma. Pediatric Blood and Cancer2013; Vol. 60, issue 5:879‐84. CENTRAL

Frgala 2007 {published data only}

Frgala T, Dubska L, Reynolds CP. Retinoids in therapy of neuroblastoma. Klinicka Onkologie 2007;20:311‐4. CENTRAL

Garaventa 2003 {published data only}

Garaventa A, Luksch R, Lo Piccolo MS, Cavadini E, Montaldo PG, Pizzitola MR, et al. Phase I trial and pharmacokinetics of fenretinide in children with neuroblastoma. Clinical Cancer Research 2003;9(6):2032‐9. CENTRAL

Granger 2012 {published data only}

Granger M, Grupp SA, Kletzel M, Kretschmar C, Naranjo A, London WB, et al. Feasibility of a tandem autologous peripheral blood stem cell transplant regimen for high risk neuroblastoma in a cooperative group setting: A Pediatric Oncology Group study: A Report from the Children's Oncology Group. Pediatric Blood and Cancer 2012;59(5):902‐7. CENTRAL

Grissom 1996 {published data only}

Grissom LE, Griffin GC, Mandell GA. Hypervitaminosis A as a complication of treatment for neuroblastoma. Pediatric Radiology 1996;26(3):200‐2. CENTRAL

Gyorfy 2003 {published data only}

Gyorfy A, Kovacs T, Szegedi I, Olah E, Kiss C. Sweet syndrome associated with 13‐cis‐retinoic acid (isotretinoin) therapy. Medical Pediatric Oncology 2003;40(2):135‐6. CENTRAL

Hamidieh 2012 {published data only}

Hamidieh AA, Beigi D, Fallahi B, Behfar M, Jalili M, Hamdi A, et al. Comparison of autologous hematopoietic stem cell transplantation with and without metaiodobenzylguanidine (MIBG) in patients with high risk neuroblastoma. 2012 BMT Tandem Meetings San Diego, CA United States. Biology of Blood and Marrow Transplantation 2012;18(2 Suppl):S251. CENTRAL

Haysom 2005 {published data only}

Haysom L, Ziegler DS, Cohn RJ, Rosenberg AR, Carroll SL, Kainer G. Retinoic acid may increase the risk of bone marrow transplant nephropathy. Pediatric Nephrology 2005;20(4):534‐8. CENTRAL

Hoefer‐Janker 1969 {published data only}

Hoefer‐Janker H, Khazne F, Scheef W. 1st clinical experience with subtoxic vitamin A doses during radiation and cytostatic tumor therapy [Erste klinische Erfahrungen mit subtoxischen Vitamin‐A‐Dosen im Rahmen der radiologischen und zytostatichen Tumortherapie]. Krebsarzt 1969;24(4):203‐7. CENTRAL

Inamo 1999 {published data only}

Inamo Y, Suzuki T, Mugishima H. A case of growth failure caused by 13‐CIS‐retinoic acid administration after bone marrow transplantation for neuroblasoma. Endocrine Journal 1999;46 Suppl:S113‐5. CENTRAL

Kazanowska 2008 {published data only}

Kazanowska B, Reich A, Jelen M, Chybicka A. Chronic metastatic neuroblastoma. Pediatric Blood and Cancer 2008;50(4):898‐900. CENTRAL

Khan 1996 {published data only}

Khan AA, Villablanca JG, Reynolds CP, Avramis VI. Pharmacokinetic studies of 13‐cis‐retinoic acid in pediatric patients with neuroblastoma following bone marrow transplantation. Cancer Chemotherapy and Pharmacology 1996;39(1‐2):34‐41. CENTRAL

Kletzel 2002 {published data only}

Kletzel M, Katzenstein HM, Haut PR, Yu AL, Morgan E, Reynolds M, et al. Treatment of high‐risk neuroblastoma with triple‐tandem high‐dose therapy and stem‐cell rescue: Results of the Chicago Pilot II study. Journal of Clinical Oncology 2002;20(9):2284‐92. CENTRAL

Kogner 2004 {published data only}

Kogner P, Borgström P, Karpe B, Lundell G, Hjelm Skog AL, Winiarski J, et al. Children with high‐risk neuroblastoma may be long‐term survivors after application of intensified multimodal therapy. 36th Congress of the International Society of Paediatric Oncology, SIOP 2004, Oslo, Norway, Abstract P.C.005. Pediatric Blood and Cancer2004; Vol. 43, issue 4:400. CENTRAL

Kohler 2000 {published data only}

Kohler JA, Imeson J, Ellershaw C, Lie SO. A randomized trial of 13‐Cis retinoic acid in children with advanced neuroblastoma after high‐dose therapy. British Journal of Cancer 2000;83(9):1124‐7. CENTRAL

Kreissman 2013 {published data only}

Kreissman SG, Seeger RC, Matthay KK, London WB, Sposto R, Grupp SA, et al. Purged versus non‐purged peripheral blood stem‐cell transplantation for high‐risk neuroblastoma (COG A3973): a randomised phase 3 trial. Lancet Oncology 2013;14(10):999‐1008. CENTRAL

Kushner 1994 {published data only}

Kushner BH, LaQuaglia MP, Bonilla MA, Lindsley K, Rosenfield N, Yeh S, et al. Highly effective induction therapy for stage 4 neuroblastoma in children over 1 year of age. Journal of Clinical Oncology 1994;12(12):2607‐13. CENTRAL

Kushner 2001a {published data only}

Kushner BH, Kramer K, Cheung NK. Phase II trial of the anti‐GD2 monoclonal antibody 3F8 and granulocyte‐macrophage colony‐stimulating factor for neuroblastoma. Journal of Clinical Oncology 2001;19(22):4189‐94. CENTRAL

Kushner 2001b {published data only}

Kushner BH, Cheung NK, Kramer K, Dunkel IJ, Calleja E, Boulad F. Topotecan combined with myeloablative does of thiotepa and carboplatin for neuroblastoma, brain tumors, and other poor‐risk solid tumors in children and young adults. Bone Marrow Transplantation 2001;28(6):551‐6. CENTRAL

Kushner 2003a {published data only}

Kushner BH, Kramer K, LaQuaglia MP, Cheung NK. Curability of recurrent disseminated disease after surgery alone for local‐regional neuroblastoma using intensive chemotherapy and anti‐G(D2) immunotherapy. Journal of Pediatric Hematology and Oncology 2003;25(7):515‐9. CENTRAL

Kushner 2003b {published data only}

Kushner BH, Kramer K, LaQuaglia MP, Modak S, Cheung NK. Neuroblastoma in adolescents and adults: the Memorial Sloan‐Kettering experience. Medical Pediatric Oncology 2003;41(6):508‐15. CENTRAL

Kushner 2015 {published data only}

Kushner BH, Ostrovnaya I, Cheung IY, Kuk D, Kramer K, Modak S, et al. Prolonged progression‐free survival after consolidating second or later remissions of neuroblastoma with Anti‐GD2 immunotherapy and isotretinoin: A prospective Phase II study. Oncoimmunology 2015;4(7):e1016704. CENTRAL

Kushner 2016 {published data only}

Kushner BH, Ostrovnaya I, Cheung IY, Kuk D, Modak S, Kramer K, et al. Lack of survival advantage with autologous stem‐cell transplantation in high‐risk neuroblastoma consolidated by anti‐GD2 immunotherapy and isotretinoin. Oncotarget 2016;7(4):4155‐66. CENTRAL

Ladenstein 2004 {published data only}

Ladenstein R, Pötschger U, Modritz D, Schreier G, Castel V, Michon J, et al. The siopen‐r‐net project: building a european network for neuroblastoma treatment (hr‐nbl‐1/esiop trial) and research. 36th Congress of the International Society of Paediatric Oncology, SIOP 2004, Oslo, Norway, Abstract P.C.014. Pediatric Blood and Cancer 2004;43(4):402. CENTRAL

Ladenstein 2014 {published data only}

Ladenstein RL, Poetschger U, Luksch R, Brock P, Castel V, Yaniv I, et al. Immunotherapy (IT) with ch14.18/CHO for high‐risk neuroblastoma: First results from the randomised HR‐NBL1/SIOPEN trial. 2014 ASCO Annual Meeting. Journal of Clinical Oncology 2014;32(5 suppl):abstr 10026. CENTRAL

Laskin 2011 {published data only}

Laskin BL, Goebel J, Davies SM, Khoury JC, Bleesing JJ, Mehta PA, et al. Early clinical indicators of transplant‐associated thrombotic microangiopathy in pediatric neuroblastoma patients undergoing auto‐SCT. Bone Marrow Transplantation 2011;46(5):682‐9. CENTRAL

Levin 2006 {published data only}

Levin VA, Giglio P, Puduvalli VK, Jochec J, Groves MD, Yung WK, et al. Combination chemotherapy with 13‐cis‐retinoic acid and celecoxib in the treatment of glioblastoma multiforme. Journal of Neurooncology 2006;78(1):85‐90. CENTRAL

Lie 1993 {published data only}

Lie SO. Retinoids in the treatment of neuroblastoma. [abstract no: 204 ]. European Journal of Cancer. 1993; Vol. 29a(Suppl 6):S42. CENTRAL

Marabelle 2009 {published data only}

Marabelle A, Sapin V, Rousseau R, Periquet B, Demeocq F, Kanold J. Hypercalcemia and 13‐cis‐retinoic acid in post‐consolidation therapy of neuroblastoma. Pediatric Blood and Cancer 2009;52(2):280‐3. CENTRAL

Maris 2000 {published data only}

Maris JM, Weiss MJ, Guo C, Gerbing RB, Stram DO, White PS, et al. Loss of heterozygosity at 1p36 independently predicts for disease progression but not decreased overall survival probability in neuroblastoma patients: A Children's Cancer Group study. Journal of Clinical Oncology 2000;18(9):1888‐99. CENTRAL

Marmor 2008 {published data only}

Marmor MF, Jain A, Moshfeghi D. Total rod ERG suppression with high dose compassionate Fenretinide usage. Documenta Ophthalmologica 2008;117(3):257‐61. CENTRAL

Mastrangelo 2011 {published data only}

Mastrangelo S, Rufini V, Ruggiero A, Di GA, Riccardi R. Treatment of advanced neuroblastoma in children over 1 year of age: The critical role of 131I‐metaiodobenzylguanidine combined with chemotherapy in a rapid induction regimen. Pediatric Blood and Cancer 2011;56(7):1032‐40. CENTRAL

Matthay 1995 {published data only}

Matthay KK, O'Leary MC, Ramsay NK, Villablanca J, Reynolds CP, Atkinson JB, et al. Role of myeloablative therapy in improved outcome for high risk neuroblastoma: Review of recent Children's Cancer Group results. European Journal of Cancer 1995;31A(4):572‐5. CENTRAL

Matthay 1999a {published data only}

Matthay KK. Current results with myeloablative therapy with hematopoietic support in advanced neuroblastoma. Cancer Research Therapy and Control 1999;9:89‐94. CENTRAL

Matthay 2000 {published data only}

Matthay KK, Reynolds CP. Is there a role for retinoids to treat minimal residual disease in neuroblastoma?. British Journal of Cancer 2000;83(9):1121‐3. CENTRAL

Matthay 2006 {published data only}

Matthay KK, Tan JC, Villablanca JG, Yanik GA, Veatch J, Franc B, et al. Phase I dose escalation of iodine‐131‐metaiodobenzylguanidine with myeloablative chemotherapy and autologous stem‐cell transplantation in refractory neuroblastoma: a new approaches to Neuroblastoma Therapy Consortium Study. Journal of Clinical Oncology 2006;24(3):500‐6. CENTRAL

Matthay 2013 {published data only}

Matthay KK. Targeted isotretinoin in neuroblastoma: kinetics, genetics, or absorption. Clinical Cancer Research 2013;19(2):311‐3. CENTRAL

Maurer 2013 {published data only}

Maurer BJ, Kang MH, Villablanca JG, Janeba J, Groshen S, Matthay KK, et al. Phase I trial of fenretinide delivered orally in a novel organized lipid complex in patients with relapsed/refractory neuroblastoma: a report from the new approaches to neuroblastoma therapy (NANT) consortium. Pediatric Blood and Cancer 2013;60(11):1801‐8. CENTRAL

Maurer 2014 {published data only}

Maurer BJ, Bender JLG, Kang MH, Villablanca J, Wei D, Groshen SG, et al. Fenretinide (4‐HPR)/Lym‐X‐Sorb (LXS) oral powder plus ketoconazole in patients with high‐risk (HR) recurrent or resistant neuroblastoma: A new approach to neuroblastoma therapy (NANT) consortium trial. 2014 ASCO Annual Meeting. Journal of Clinical Oncology 2014;32(5 suppl):abstr 10071. CENTRAL

McCann 1993 {published data only}

McCann J. ASCO/AACR: shared emphases and separate meetings. Journal of the National Cancer Institute 1993;85(12):939‐42. CENTRAL

Mora 2015 {published data only}

Mora J, Cruz O, Lavarino C, Rios J, Vancells M, Parareda A. Results of induction chemotherapy in children older than 18 months with stage‐4 neuroblastoma treated with an adaptive‐to‐response modified N7 protocol (mN7). 2015 Clinical and Translational Oncology;17(7):521‐9. CENTRAL

Mostoufi‐Moab 2016 {published data only}

Mostoufi‐Moab S. Skeletal impact of retinoid therapy in childhood cancer survivors. Pediatric Blood & Cancer 2016;63(11):1884‐5. CENTRAL

Mugishima 1995 {published data only}

Mugishima H, Harada K, Suzuki T, Chin M, Shimada T, Takamura M, et al. Comprehensive treatment of advanced neuroblastoma involving autologous bone marrow transplant. Acta Paediatrica Japonica 1995;37(4):493‐9. CENTRAL

Mugishima 2008 {published data only}

Mugishima H, Chin M, Suga M, Schichino H, Ryo N, Nakamura M, et al. Hypercalcemia induced by 13 cis‐retinoic acid in patients with neuroblastoma. Pediatrics International 2008;50(2):235‐7. CENTRAL

Nishimura 1997 {published data only}

Nishimura G, Mugishima H, Hirao J, Yamato M. Generalized metaphyseal modification with cone‐shaped epiphyses following long‐term administration of 13‐cis‐retinoic acid. European Journal of Pediatrics 1997;156(6):432‐5. CENTRAL

Olgun 2008 {published data only}

Olgun N, Gunes D, Aksoylar S, Varan A, Erbay A, Hazar V, et al. The Turkish pediatric oncology group neuroblastoma 2003 (tpog‐nb‐2003): treatment results of the high risk group. 40th Annual Conference of the International Society of Paediatric Oncology, SIOP 2008, Berlin, Germany, Abstract D110. Pediatric Blood and Cancer 2008;50(5 Suppl):140. CENTRAL

Ozkaynak 2014 {published data only}

Ozkaynak MF, Gilman AL, Yu AL, London WB, Sondel PM, Smith MA, et al. A comprehensive safety trial of chimeric antibody 14.18 (ch14.18) with GM‐CSF, IL‐2, and isotretinoin in high‐risk neuroblastoma patients following myeloablative therapy: A Children's Oncology Group study. 2014 ASCO Annual Meeting. Journal of Clinical Oncology 2014;32(5 suppl):abstr 10044. CENTRAL

Park 2005 {published data only}

Park JR, Villablanca JG, Seeger R, Shimada H, London W, Gerbing R, et al. Outcome of high risk (HR) stage 3 neuroblastoma (NB) with myeloablative therapy and 13‐cis‐retinoic acid. Abstracts for the 41th Annual Meeting of the American Society of Clinical Oncology, Orlando,FL, 13‐17 May, 2005 [Abstract No. 8503]. Annual Meeting Proceedings of the American Society of Clinical Oncology. Alexandria: American Society of Clinical Oncology, 2005; Vol. 23:800. CENTRAL

Park 2011 {published data only}

Park JR, Scott JR, Stewart CF, London WB, Naranjo A, Santana VM, et al. Pilot induction regimen incorporating pharmacokinetically guided topotecan for treatment of newly diagnosed high‐risk neuroblastoma: a Children's Oncology Group study. Journal of Clinical Oncology 2011;29(33):4351‐7. CENTRAL

Pearson 2008 {published data only}

Pearson AD, Pinkerton CR, Lewis IJ, Imeson J, Ellershaw C, Machin D. High‐dose rapid and standard induction chemotherapy for patients aged over 1 year with stage 4 neuroblastoma: a randomised trial. Lancet Oncology2008; Vol. 9, issue 3:247‐56. CENTRAL

Rayburg 2009 {published data only}

Rayburg M, Towbin A, Yin H, Maugans T, Maurer B, Nagarajan R, et al. Langerhans cell histiocytosis in a patient with stage 4 neuroblastoma receiving oral fenretinide. Pediatric Blood and Cancer 2009;53(6):1111‐3. CENTRAL

Reed 1999 {published data only}

Reed JC. Fenretinide: the death of a tumor cell. Journal of the National Cancer Institute 1999;91(13):1099‐100. CENTRAL

Reynolds 2001 {published data only}

Reynolds CP, Seeger RC. Detection of minimal residual disease in bone marrow during or after therapy as a prognostic marker for high‐risk neuroblastoma. Journal of Pediatric Hematology/Oncology 2001;23(3):150‐2. CENTRAL

Richtig 2005 {published data only}

Richtig E, Soyer HP, Posch M, Mossbacher U, Bauer P, Teban L, et al. Prospective, randomized, multicenter, double‐blind placebo‐controlled trial comparing adjuvant interferon alfa and isotretinoin with interferon alfa alone in stage IIA and IIB melanoma: European Cooperative Adjuvant Melanoma Treatment Study Group. Journal of Clinical Oncology2005; Vol. 23, issue 34:8655‐63. CENTRAL

Rustin 1982 {published data only}

Rustin GJ, Bagshawe KD. Trial of an aromatic retinoid in patients with solid tumours. British Journal of Cancer 1982;45(2):304‐8. CENTRAL

Saarinen‐Pihkala 2012 {published data only}

Saarinen‐Pihkala UM, Hovi L, Koivusalo A, Jahnukainen K, Karikoski R, Sariola H, et al. Thiotepa and melphalan based single, tandem, and triple high dose therapy and autologous stem cell transplantation for high risk neuroblastoma. Pediatric Blood and Cancer 2012;59(7):1190‐7. CENTRAL

Sato 2012 {published data only}

Sato Y, Kuwashima S, Kurosawa H, Sugita K, Fukushima K, Arisaka O. 13‐cis‐retinoic acid‐associated bone marrow edema in neuroblastoma. Pediatric Blood and Cancer 2012;59(3):589‐90. CENTRAL

Seeger 2000 {published data only}

Seeger RC, Reynolds CP, Gallego R, Stram DO, Gerbing RB, Matthay KK. Quantitative tumor cell content of bone marrow and blood as a predictor of outcome in stage IV neuroblastoma: a Children's Cancer Group Study. Journal of Clinical Oncology 2000;18(24):4067‐76. CENTRAL

Simon 2005 {published data only}

Simon T, Hero B, Faldum A, Handgretinger R, Schrappe M, Niethammer D, et al. Infants with stage 4 neuroblastoma: the impact of the chimeric anti‐GD2‐antibody ch14.18 consolidation therapy. Klinische Pädiatrie 2005;217(3):147‐52. CENTRAL

Simon 2011 {published data only}

Simon T, Hero B, Handgretinger R, Schrappe M, Klingebiel T, Fruehwald M, et al. Anti‐GD2‐antibody CH14.18 or retinoic acid as consolidation therapy in high‐risk neuroblastoma. 43rd Congress of the International Society of Paediatric Oncology, SIOP 2011 Auckland New Zealand. Pediatric Blood and Cancer. 2011; Vol. 57, issue 5:789. CENTRAL

Sirachainan 2008 {published data only}

Sirachainan N, Visudtibhan A, Tuntiyatorn L, Pakakasama S, Chuansumrit A, Hongeng S. Favorable response of intraommaya topotecan for leptomeningeal metastasis of neuroblastoma after intravenous route failure. Pediatric Blood and Cancer 2008;50(1):169‐72. CENTRAL

Sung 2007 {published data only}

Sung KW, Lee SH, Yoo KH, Jung HL, Cho EJ, Koo HH, et al. Tandem high‐dose chemotherapy and autologous stem cell rescue in patients over 1 year of age with stage 4 neuroblastoma. Bone Marrow Transplantation 2007;40(1):37‐45. CENTRAL

Tang 2006 {published data only}

Tang JY, Pan C, Chen J, Xu M, Chen J, Xue HL, et al. Comprehensive protocol for diagnosis and treatment of childhood neuroblastoma‐‐results of 45 cases [Chinese language]. Zhonghua Er Ke Za Zhi (Chinese Journal of Pediatrics) 2006;44(10):770‐3. CENTRAL

Turman 1999 {published data only}

Turman MA, Hammond S, Grovas A, Rauck AM. Possible association of retinoic acid with bone marrow transplant nephropathy. Pediatric Nephrology 1999;13(9):755‐8. CENTRAL

Veal 2007 {published data only}

Veal GJ, Cole M, Errington J, Pearson AD, Foot AB, Whyman G, et al. Pharmacokinetics and metabolism of 13‐cis‐retinoic acid (isotretinoin) in children with high‐risk neuroblastoma ‐ a study of the United Kingdom Children's Cancer Study Group. British Journal of Cancer 2007;96(3):424‐31. CENTRAL

Veal 2013 {published data only}

Veal GJ, Errington J, Rowbotham SE, Illingworth NA, Malik G, Cole M, et al. Adaptive dosing approaches to the individualization of 13‐cis‐retinoic acid (isotretinoin) treatment for children with high‐risk neuroblastoma. Clinical Cancer Research 2013;19(2):469‐79. CENTRAL

Villablanca 1992 {published data only}

Villablanca JG, Avramis VI, Khan AA, Matthay KK, Ramsay NKC, Seeger RC, et al. Phase I trial of 13‐Cis‐Retinoic acid (cis‐RA) in neuroblastoma patients following bone marrow transplantation (BMT) [abstract]. Proceedings of the American Society of Clinical Oncology. Alexandria: American Society of Clinical Oncology, 1992; Vol. 11:366, Abstract 1263. CENTRAL

Villablanca 1993 {published data only}

Villablanca JG, Khan AA, Avramis VI, Reynolds CP. Hypercalcemia: a dose‐limiting toxicity associated with 13‐cis‐retinoic acid. American Journal of Pediatric Hematology/Oncology 1993;15(4):410‐5. CENTRAL

Villablanca 1995 {published data only}

Villablanca JG, Khan AA, Avramis VI, Seeger RC, Matthay KK, Ramsay NK, et al. Phase I trial of 13‐cis‐retinoic acid in children with neuroblastoma following bone marrow transplantation. Journal of Clinical Oncology 1995;13(4):894‐901. CENTRAL

Villablanca 2006 {published data only}

Villablanca JG, Krailo MD, Ames MM, Reid JM, Reaman GH, Reynolds CP. Phase I trial of oral fenretinide in children with high‐risk solid tumors: a report from the Children's Oncology Group (CCG 09709). Journal of Clinical Oncology 2006;24(21):3423‐30. CENTRAL

Villablanca 2011 {published data only}

Villablanca JG, London WB, Naranjo A, McGrady P, Ames MM, Reid JM, et al. Phase II study of oral capsular 4‐hydroxyphenylretinamide (4‐HPR/fenretinide) in pediatric patients with refractory or recurrent neuroblastoma: a report from the Children's Oncology Group. Clincal Cancer Research 2011;17(21):6858‐66. CENTRAL

Weitman 1997 {published data only}

Weitman S, Ochoa S, Sullivan J, Shuster J, Winick N, Pratt C, et al. Pediatric phase II cancer chemotherapy trials: A Pediatric Oncology Group study. Journal of Pediatric Hematology/Oncology1997; Vol. 19, issue 3:187‐91. CENTRAL

Ye 2010 {published data only}

Ye QD, Tang JY, Pan C, Chen J, Xue HL, Chen J, et al. Therapeutic experience of childhood stage III neuroblastoma [Chinese language]. Zhonghua Yi Xue Za Zhi 2010;90(22):1556‐8. CENTRAL

Yu 2010 {published data only}

Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, et al. Anti‐GD2 antibody with GM‐CSF, interleukin‐2, and isotretinoin for neuroblastoma. New England Journal of Medicine2010; Vol. 363, issue 14:1324‐34. CENTRAL

Yung 1996 {published data only}

Yung WK, Kyritsis AP, Gleason MJ, Levin VA. Treatment of recurrent malignant gliomas with high‐dose 13‐cis‐retinoic acid. Clinical Cancer Research 1996;2(12):1931‐5. CENTRAL

Zhu 2010 {published data only}

Zhu X, Huang D, Zhang W, Wang Y, Song Q, Zhang Y, et al. Therapeutic effects of high dose chemotherapy combined with autologous peripheral blood stem cell transplantation on 18 cases of stage IV neuroblastoma in children. Chinese Journal of Clinical Oncology 2010;37:467‐70. CENTRAL

References to studies awaiting assessment

Reynolds 1998 {published data only}

Reynolds CP, Villablanca JG, Stram DO, Harris R, Seeger RC, Matthay KK. 13‐CIS‐retinoic acid after intensive consolidation therapy for neuroblastoma improves event‐free survival: a randomized Children's Cancer Group study. 34th Annual Meeting of the American Society of Clinical Oncology; 16‐19 May 1998; Los Angeles, California, USA. Proceedings of the American Society of Clinical Oncology. Alexandria: American Society of Clinical Oncology, 1998; Vol. 17:2a, Abstract 5. CENTRAL

Reynolds 2002 {published data only}

Reynolds CP, Villablanca JG, Gerbing RB, Stram DO, Seeger RC, Matthay KK. 13‐cis‐retinoic acid improves overall survival following myeloablative therapy for high‐risk neuroblastoma: a randomized Children's Cancer Group study. 38th Annual Meeting of the American Society of Clinical Oncology; 18‐21 May 2002; Orlando, Florida, USA. Proceedings of the American Society of Clinical Oncology. Alexandria: American Society of Clinical Oncology, 2002; Vol. 21 (Pt 1):392a, Abstract 1564. CENTRAL

Berthold 2005

Berthold F, Simon T. Clinical presentation. In: Cheung NKV, Cohn SL editor(s). Neuroblastoma. Berlin: Springer, 2005:63‐85.

Brisse 2011

Brisse HJ, McCarville MB, Granata C, Krug KB, Wootton‐Gorges SL, Kanegawa K, et al. Guidelines for imaging and staging of neuroblastic tumors: consensus report from the International Neuroblastoma Risk Group Project. Radiology 2011;261(1):243‐57.

Brodeur 1993

Brodeur GM, Pritchard J, Berthold F, Carlsen NL, Castel V, Castelberry RP, et al. Revisions of the international criteria for neuroblastoma diagnosis, staging, and response to treatment. Journal of Clinical Oncology 1993;11(8):1466‐77.

CDC 2000a

Centers of Disease Control and Prevention (CDC). Clinical growth charts. Birth to 36 months: boys length‐for‐age and weight‐for‐age percentiles. www.cdc.gov/growthcharts/data/set1clinical/cj41c017.pdf (accessed 18 May 2017).

CDC 2000b

Centers of Disease Control and Prevention. Clinical growth charts. 2 to 20 years: boys stature‐for‐age and weight‐for‐age percentiles. www.cdc.gov/growthcharts/data/set1clinical/cj41c021.pdf (accessed 18 May 2017).

Cheung 2014

Cheung NK, Modak S, Ostranova I, Roberts SS, Basu EM, Kramer K, et al. When overall survival fails to confirm event‐free survival, should the latter be used to set the standard of care?. Journal of Clinical Oncology 2014;32(36):4173‐4.

Cohn 2009

Cohn SL, Pearson AD, London WB, Monclair T, Ambros PF, Brodeur GM, et al. The International Neuroblastoma Risk Group (INRG) classification system: an INRG Task Force report. Journal of Clinical Oncology 2009;27(2):289‐97.

Cole 2012

Cole KA, Maris JM. New strategies in refractory and recurrent neuroblastoma: translational opportunities to impact patient outcome. Clininical Cancer Research 2012;18(9):2423‐8.

CTEP 2010

Cancer Therapy Evaluation Program (CTEP). Common Terminology Criteria for Adverse Events (CTCAE). ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm#ctc_40 (accessed 18 May 2017).

DerSimonian 1986

DerSimonian R, Laird N. Meta‐analysis in clinical trials. Controlled Clinical Trials 1986;7(3):177‐88.

Erratum 2014

Errata. Journal of Clinical Oncology2014; Vol. 32, issue 17:1862‐3. CENTRAL

Esiashvili 2007

Esiashvili N, Goodman M, Ward K, Marcus RB, Johnstone PA. Neuroblastoma in adults: Incidence and survival analysis based on SEER data. Pediatric Blood & Cancer 2007;49(1):41‐6.

Evans 1971

Evans AE, D'Angio GJ, Randolph J. A proposed staging for children with neuroblastoma. Children's Cancer Study Group A. Cancer 1971;27(2):374‐8.

GARD 2017

Genetic and Rare Diseases Information Center (GARD), The Office of Rare Diseases Research (ORDR). Neuroblastoma. rarediseases.info.nih.gov/gard/7185/neuroblastoma/resources/1 (accessed 18 May 2017).

Goodman 2012

Goodman MT, Gurney JG, Smith MA, Olshan AF. SEER Pediatric Monograph. Sympathetic Nervous System Tumors. seer.cancer.gov/publications/childhood/sympathetic.pdf (accessed 18 May 2017).

GRADEpro 2015 [Computer program]

McMaster University (developed by Evidence prime). GRADEpro GDT. Version accessed 18 May 2017. Hamilton (ON): McMaster University (developed by Evidence prime), 2015.

Hero 2008

Hero B, Simon T, Spitz R, Ernestus K, Gnekow AK, Scheel‐Walter HG, et al. Localized infant neuroblastomas often show spontaneous regression: results of the prospective trials NB95‐S and NB97. Journal of Clinical Oncology 2008;26(9):1504‐10.

Higgins 2003

Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ 2003;327(7414):557‐60.

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 www.handbook.cochrane.org.

ICSSC 2003

International Clinical Sciences Support Center (ICSSC). WHO Toxicity Grading Scale for Determining The Severity of Adverse Events. www.icssc.org/Documents/Resources/AEManual2003AppendicesFebruary_06_2003%20final.pdf (accessed 18 May 2017).

Kremer 2008

Kremer LCM, Van Dalen EC, Moher D, Caron HN. Cochrane Childhood Cancer Group. About The Cochrane Collaboration (Cochrane Review Groups (CRGs)). onlinelibrary.wiley.com/o/cochrane/clabout/articles/CHILDCA/frame.html (accessed 18 May 2017).

Matthay 2012

Matthay KK, George RE, Yu AL. Promising therapeutic targets in neuroblastoma. Clinical Cancer Research 2012;18(10):2740‐53.

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group. Preferred reporting items for systematic reviews and meta‐analyses: the PRISMA statement. PLoS Medicine 2009;6(7):e1000097.

NCI PDQ 2017

National Cancer Institute: PDQ® Neuroblastoma Treatment. Bethesda, MD: National Cancer Institute. Updated 14 April 2017. http://cancer.gov/cancertopics/pdq/treatment/neuroblastoma/HealthProfessional (accessed 18 May 2017).

Parmar 1998

Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta‐analyses of the published literature for survival endpoints. Statistics in Medicine 1998;17(24):2815‐34.

Ratko 2012

Ratko TA, Belinson SE, Brown HM, Noorani HZ, Chopra RD, Marbella A, et al. Hematopoietic stem‐cell transplantation in the pediatric population. AHRQ Comparative Effectiveness Reviews. www.ncbi.nlm.nih.gov/books/NBK84626/ (accessed 18 May 2017).

Review Manager 2014 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.3. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Reynolds 2003

Reynolds CP, Matthay KK, Villablanca JG, Maurer BJ. Retinoid therapy of high‐risk neuroblastoma. Cancer Letters 2003;197(1‐2):185‐92.

Schünemann 2011

Schünemann HJ, Oxman AD, Vist GE, Higgins JPT, Deeks JJ, Glasziou P, et al. Chapter 12: Interpreting results and drawing conclusions. In: 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 www.handbook.cochrane.org.

Shimada 1984

Shimada H, Chatten J, Newton WA, Sachs N, Hamoudi AB, Chiba T, et al. Histopathologic prognostic factors in neuroblastic tumors: definition of subtypes of ganglioneuroblastoma and an age‐linked classification of neuroblastomas. Journal of the National Cancer Institute 1984;73(2):405‐16.

Sidell 1982

Sidell N. Retinoic acid‐induced growth inhibition and morphologic differentiation of human neuroblastoma cells in vitro. Journal of the National Cancer Institute 1982;68(4):589‐96.

Sidell 1983

Sidell N, Altman A, Haussler MR, Seeger RC. Effects of retinoic acid (RA) on the growth and phenotypic expression of several human neuroblastoma cell lines. Experimental Cell Research 1983;148(1):21–30.

Tierney 2007

Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR. Practical methods for incorporating summary time‐to‐event data into meta‐analysis. Trials 2007;8:16.

Yalçin 2015

Yalçin B, Kremer LCM, Van Dalen EC. High‐dose chemotherapy and autologous haematopoietic stem cell rescue for children with high‐risk neuroblastoma. Cochrane Database of Systematic Reviews 2015, Issue 10. [DOI: 10.1002/14651858.CD006301.pub4]

Yoon 2012

Yoon U, Knobloch K. Assessment of reporting quality of conference abstracts in sports injury prevention according to CONSORT and STROBE criteria and their subsequent publication rate as full papers. BMC Medical Research Methodology 2012;12:47.

References to other published versions of this review

Peinemann 2013

Peinemann F, Bartel C, Grouven U, Berthold F. Retinoic acid post consolidation therapy for high‐risk neuroblastoma. Cochrane Database of Systematic Reviews 2013, Issue 7. [DOI: 10.1002/14651858.CD010685]

Peinemann 2015

Peinemann F, Van Dalen EC, Tushabe DA, Berthold F. Retinoic acid post consolidation therapy for high‐risk neuroblastoma patients treated with autologous hematopoietic stem cell transplantation. Cochrane Database of Systematic Reviews 2015, Issue 1. [DOI: 10.1002/14651858.CD010685.pub2]

Peinemann 2016

Peinemann F, Van Dalen EC, Berthold F. Retinoic acid for high‐risk neuroblastoma patients after autologous stem cell transplantation ‐ Cochrane Review. Klinische Padiatrie 2016;228(3):124‐9.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Matthay 1999

Methods

Setting

  • Multicentre study denoted CCG‐3891. The abbreviation CCG derives from the Children's Cancer Group that merged in 2000 with other groups to form the Children's Oncology Group

  • United States

Duration of enrollment

  • From 1991 to 1996

Randomisation

  • A permuted‐block design was used for the random assignment of approximately equal numbers of participants from each of 2 strata (those with and those without metastatic disease)

  • The study design included 2 randomisations. The 2nd randomisation was similarly balanced with respect to the numbers of participants from each group of the first randomisation and non‐randomised patients who were ineligible for transplantation

  • First randomisation: Participants without progressive disease after 2 of 5 cycles of initial chemotherapy were randomly allocated to cytotoxic therapy group 1 that consisted of myeloablative therapy and autologous bone marrow transplantation or to cytotoxic therapy group 2 that consisted of 3 cycles of continuation chemotherapy. Participants with progressive disease were non‐randomly assigned to continuation chemotherapy

  • 2nd randomisation: Participants without progressive disease after consolidation therapy were randomly assigned to 13‐cis‐retinoic acid or to no further therapy

Median follow‐up time

  • After diagnosis: median of 43 months (range 2 to 89) for 539 participants eligible for initial chemotherapy; not reported separately for the 98 participants eligible for this review

  • From transplantation to start of 13‐cis‐retinoic acid median of 97 days; no information on time from transplantation to 2nd randomisation for the 98 participants eligible for this review

  • Length of follow‐up from 2nd randomisation until end of study not reported for the 98 participants eligible for this review

Participants

Eligibility criteria

  • Newly‐diagnosed high‐risk neuroblastoma patients; high‐risk neuroblastoma was defined as: stage IV neuroblastoma; stage III disease with 1 or more of the following: amplification of the MYCN oncogene, a serum ferritin level of at least 143 ng per mL, and unfavourable histopathological findings; stage II disease with amplification of MYCN (age > 1 year); stage I or II disease with bone metastases before therapy other than surgery; and stage IV disease with MYCN amplification for < 1 year. Staging was done using the Evans staging criteria (Evans 1971). Unfavourable histopathological findings were based on the Shimada classification (Shimada 1984)

  • 1 to 18 years of age

Number of patients eligible for this review

  • 98 participants after high‐dose chemotherapy followed by bone marrow transplantation: 50 received retinoic acid and 48 received no further therapy

Age

  • Median and range not reported for 98 participants eligible for this review

Gender

  • Not reported

Stage of disease

  • Not reported for 98 participants eligible for this review

Remission status

  • None of the 98 participants eligible for this review had progressive disease at the time of the 2nd randomisation; no further information provided

Compliance with randomisation

  • Not reported for the 2nd randomisation

Previous treatment, except initial and consolidation chemotherapy

  • Not reported

Comorbidity

  • Not reported

Interventions

All participants

  • Initial chemotherapy for 5 cycles at 28‐day intervals. 1 cycle consisted of: cisplatin (60 mg/m2 total dose), doxorubicin (30 mg/m2 total dose), etoposide (200 mg/m2 total dose), and cyclophosphamide (2000 mg/m2 total dose). Following induction chemotherapy patients with gross residual disease received surgery and radiotherapy (number of participants not reported)

First randomisation

BMT arm (patients in the continuation chemotherapy arm were not eligible for this review)

  • Carboplatin (1000 mg/m2 total dose), etoposide (640 mg/m2 total dose), melphalan (210 mg/m2 total dose), total‐body irradiation (1000 cGy), purged bone marrow harvested at end of cycle 2, and granulocyte‐macrophage colony‐stimulating factor (250 microgram/m2 per day, number of days not reported)

2nd randomisation

Retinoic acid arm

  • 50 of 98 participants after high‐dose chemotherapy followed by autologous bone marrow transplantation were randomised to the retinoic acid arm to receive 6 cycles of retinoic acid. 1 cycle consists of: 13‐Cis retinoic acid at a dose of 160 mg/mg/m2 per day for 14 consecutive days in a 28‐day cycle (14 days on, 14 days off), resulting in a cumulative dose after 28 days of 2240 mg/m2

Control arm

  • 48 of 98 participants after high‐dose chemotherapy followed by autologous bone marrow transplantation were randomised to the no‐further‐therapy arm

Outcomes

Primary outcomes

  • Overall survival (definition not provided, but measured from 2nd randomisation)

Secondary outcomes

  • Event‐free survival. According to Matthay 1999: "The primary end point, prespecified by the protocol, was event‐free survival calculated from the time of second randomisation. The events considered were relapse, disease progression, death from any cause, and a second neoplasm, whichever occurred first."

Notes

  • No competing interest reported, funding, grants, and awards received from not‐for‐profit organizations.

  • Supported in part by the National Cancer Institute; Neil Bogart Memorial Laboraties of the T.J. Martell Foundation for Leukemia, Cancer, and AIDS Research; American Institute for Cancer Research.

  • The authors of Matthay 1999 concluded in their follow‐up paper in 2009: "Myeloablative therapy and autologous hematopoietic cell rescue result in significantly better 5‐year EFS and OS than nonmyeloablative chemotherapy; cis‐RA given after consolidation independently results in significantly improved OS." We have questioned the results of the statistical analyses and the authors eventually initiated a revision and published an erratum (Erratum 2014) and revised their conclusion as follows: "Myeloablative therapy and autologous hematopoietic cell rescue result in significantly better 5‐year EFS than nonmyeloablative chemotherapy; neither myeloablative therapy with autologous hematopoietic cell rescue nor cis‐RA given after consolidation therapy significantly improved OS.”

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A permuted‐block design was used for the random assignment of approximately equal numbers of participants from each of 2 strata (those with and those without metastatic disease) to transplantation or continuation chemotherapy. The 2nd randomisation was similarly balanced with respect to the numbers of participants from each group of the first randomisation and non‐randomised participants who were ineligible for transplantation

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Blinding of participants, physicians and nurses was not reported

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The study committee and investigators were unaware of participants' treatment assignments, and the study was monitored by an independent committee according to a group sequential monitoring plan

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was not reported if all reported outcomes for all participants were assessed

Selective reporting (reporting bias)

Low risk

Reporting was in agreement with the protocol with regard to the outcome measures.

We were concerned with the possibility that data for the same participants were included in several publications. However, we did not include duplicate data in the review

Other bias

Unclear risk

Analysis was conducted using the data of the randomised participants, which should be consistent with the ITT principle. It was unclear if all 98 participants received the treatment to which they were randomised. Also, not all participants treated with transplantation in the first randomisation and without progressive disease afterwards were included in the 2nd randomisation. It is unclear how many eligible patients did not undergo the 2nd randomisation. The consequences of 2 randomisations in 1 study for the risk of bias are unclear.

AIDS: acquired immunodeficiency syndrome; BMT: bone marrow transplantation; cGy: centi‐Gray; CR: complete response; N: number.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Adamson 1997

Not intervention of interest: not HDCT followed by autologous HSCT

Adamson 2007

Not comparator of interest

Aksoylar 2013

Not comparator of interest

Anderson 2005

Not intervention of interest: not HDCT followed by autologous HSCT, only title/abstract available

Atra 1996

Not intervention of interest: not retinoic acid

Bagatell 2014

Not comparator of interest: all participants received retinoic acid

Bauters 2011

Not publication type of interest

Castel 2004

Not publication type of interest

Chan 2007

Not intervention of interest: not HDCT followed by autologous HSCT

Chen 2015

Not an intervention of interest: not a HDCT followed by autologous HSCT

Clarke 2003

Not comparator of interest: no comparator

Cross 2009

Not comparator of interest: no comparator

De Kraker 2008

Not comparator of interest

Di Bella 2009

Not intervention of interest: not HDCT followed by autologous HSCT, only title/abstract available

Dmitrovsky 2004

Not publication type of interest

Elimam 2006

Not intervention of interest: not retinoic acid, only title/abstract available

Finklestein 1992

Not intervention of interest: not HDCT followed by autologous HSCT

Formelli 2008

Not intervention of interest: not HDCT followed by autologous HSCT

Formelli 2010

Not intervention of interest: not HDCT followed by autologous HSCT

Fouladi 2010

Not outcome of interest or not reported separately

French 2013

Not intervention of interest: not retinoic acid

Frgala 2007

Not comparator of interest, only title/abstract available

Garaventa 2003

Not comparator of interest

Granger 2012

Not comparator of interest

Grissom 1996

Not comparator of interest: no comparator

Gyorfy 2003

Not comparator of interest: no comparator

Hamidieh 2012

Not comparator of interest: all participants received retinoic acid

Haysom 2005

Not comparator of interest: no comparator

Hoefer‐Janker 1969

Not diagnosis of interest, only title/abstract available

Inamo 1999

Not comparator of interest: no comparator

Kazanowska 2008

Not intervention of interest: not HDCT followed by autologous HSCT

Khan 1996

Not comparator of interest: pharmacokinetic study

Kletzel 2002

Not publication type of interest: not RCT

Kogner 2004

Not comparator of interest

Kohler 2000

Not intervention of interest: 28% (49/175) patients did not receive high‐dose chemotherapy and bone marrow transplantation confirmed by author inquiry and after contacting the first author of this study, it became clear that separate data on the eligible participants were not available

Kreissman 2013

Not comparator of interest

Kushner 1994

Not intervention of interest: not consolidation therapy

Kushner 2001a

Not intervention of interest: not HDCT followed by autologous HSCT

Kushner 2001b

Not publication type of interest: not RCT

Kushner 2003a

Not comparator of interest: no comparator

Kushner 2003b

Not comparator of interest: no comparator

Kushner 2015

Not publication type of interest: not RCT

Kushner 2016

Not a comparator of interest: both arms received retinoic acid

Ladenstein 2004

Not comparator of interest

Ladenstein 2014

Not comparator of interest

Laskin 2011

Not publication type of interest: not RCT

Levin 2006

Not diagnosis of interest

Lie 1993

Not publication type of interest: narrative review

Marabelle 2009

Not comparator of interest: no comparator

Maris 2000

Not comparator of interest

Marmor 2008

Not comparator of interest: no comparator

Mastrangelo 2011

Not publication type of interest: not RCT

Matthay 1995

Not publication type of interest: narrative review

Matthay 1999a

Not intervention of interest, only title/abstract available

Matthay 2000

Not publication type of interest: editorial

Matthay 2006

Not intervention of interest

Matthay 2013

Not publication type of interest: comment

Maurer 2013

Not comparator of interest

Maurer 2014

Not comparator of interest

McCann 1993

Not diagnosis of interest, only title/abstract available

Mora 2015

Not publication type of interest: not RCT

Mostoufi‐Moab 2016

Not publication type of interest: not RCT

Mugishima 1995

Not intervention of interest, only title/abstract available

Mugishima 2008

Not comparator of interest: no comparator

Nishimura 1997

not comparator of interest: no comparator

Olgun 2008

Not intervention of interest: not retinoic acid, only title/abstract available

Ozkaynak 2014

Not comparator of interest

Park 2005

Not publication type of interest: duplicate data

Park 2011

Not comparator of interest

Pearson 2008

Not comparator of interest: all participants received retinoic acid

Rayburg 2009

Not comparator of interest: no comparator

Reed 1999

Not diagnosis of interest: not high‐risk neuroblastoma

Reynolds 2001

Not publication type of interest: narrative review

Richtig 2005

Not diagnosis of interest: melanoma

Rustin 1982

Not diagnosis of interest: not neuroblastoma

Saarinen‐Pihkala 2012

Not comparator of interest

Sato 2012

Not comparator of interest: no comparator

Seeger 2000

Not outcome of interest or not reported separately: pharmacokinetics

Simon 2005

Not intervention of interest: not retinoic acid

Simon 2011

Not comparator of interest: all participants received retinoic acid

Sirachainan 2008

Not comparator of interest: no comparator

Sung 2007

Not comparator of interest

Tang 2006

Not comparator of interest

Turman 1999

Not comparator of interest: no comparator

Veal 2007

Not outcome of interest or not reported separately: pharmacokinetics

Veal 2013

Not outcome of interest or not reported separately: pharmacokinetics

Villablanca 1992

Not outcome of interest or not reported separately: pharmacokinetics

Villablanca 1993

Not comparator of interest: no comparator

Villablanca 1995

Not comparator of interest: no comparator

Villablanca 2006

Not comparator of interest: no comparator

Villablanca 2011

Not comparator of interest: retinoic acid independent of HDCT followed by autologous HSCT

Weitman 1997

Not comparator of interest: neuroblastoma not reported separately

Ye 2010

Not comparator of interest: retinoic acid independent of not HDCT followed by autologous HSCT, only title/abstract available

Yu 2010

Not comparator of interest: all participants received retinoic acid

Yung 1996

Not diagnosis of interest: glioma

Zhu 2010

Not intervention of interest: not retinoic acid, only title/abstract available

HDCT: high‐dose chemotherapy; HSCT: hematopoietic stem cell transplantation; RCT: randomised controlled trial

Characteristics of studies awaiting assessment [ordered by study ID]

Reynolds 1998

Methods

RCT

Participants

Patients with neuroblastoma

Interventions

13‐cis‐retinoic acid after intensive consolidation therapy

Outcomes

Event‐free survival

Notes

Title of an abstract of the annual meeting of the American Society of Clinical Oncology in 1998. Presumably associated with the study Matthay 1999.

Reynolds 2002

Methods

RCT

Participants

Patients with high‐risk neuroblastoma

Interventions

13‐cis‐retinoic acid following myeloablative therapy

Outcomes

Overall survival

Notes

Title of an abstract of the annual meeting of the American Society of Clinical Oncology in 2002. Presumably associated with the study Matthay 1999.

Data and analyses

Open in table viewer
Comparison 1. Retinoic acid versus no further therapy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.87 [0.46, 1.63]

Analysis 1.1

Comparison 1 Retinoic acid versus no further therapy, Outcome 1 Overall survival.

Comparison 1 Retinoic acid versus no further therapy, Outcome 1 Overall survival.

2 Event‐free survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.86 [0.50, 1.49]

Analysis 1.2

Comparison 1 Retinoic acid versus no further therapy, Outcome 2 Event‐free survival.

Comparison 1 Retinoic acid versus no further therapy, Outcome 2 Event‐free survival.

1Study flow diagram of current review version.Abbreviations. CT.gov: ClinicalTrials.gov; ICTRP: International Clinical Trials Registry Platform
Figuras y tablas -
Figure 1

1Study flow diagram of current review version.

Abbreviations. CT.gov: ClinicalTrials.gov; ICTRP: International Clinical Trials Registry Platform

Flow of patients in the Matthay 1999 study (as prepared by review author FP).
 Abbreviations. BMT: bone marrow transplantation; ContCT: continuation chemotherapy; CT: chemotherapy; HDCT: high‐dose chemotherapy; RA: retinoic acid.
Figuras y tablas -
Figure 2

Flow of patients in the Matthay 1999 study (as prepared by review author FP).
Abbreviations. BMT: bone marrow transplantation; ContCT: continuation chemotherapy; CT: chemotherapy; HDCT: high‐dose chemotherapy; RA: retinoic acid.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 3

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 4

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Forest plot of comparison: 1 Retinoic acid versus no further therapy, outcome: 1.1 Overall survival.
 Abbreviations. CI: confidence interval; IV: inverse variance; SE: standard error.
Figuras y tablas -
Figure 5

Forest plot of comparison: 1 Retinoic acid versus no further therapy, outcome: 1.1 Overall survival.
Abbreviations. CI: confidence interval; IV: inverse variance; SE: standard error.

Forest plot of comparison: 1 Retinoic acid versus no further therapy, outcome: 1.2 Event‐free survival.
 CI: confidence interval; IV: inverse variance; SE: standard error.
Figuras y tablas -
Figure 6

Forest plot of comparison: 1 Retinoic acid versus no further therapy, outcome: 1.2 Event‐free survival.
CI: confidence interval; IV: inverse variance; SE: standard error.

Comparison 1 Retinoic acid versus no further therapy, Outcome 1 Overall survival.
Figuras y tablas -
Analysis 1.1

Comparison 1 Retinoic acid versus no further therapy, Outcome 1 Overall survival.

Comparison 1 Retinoic acid versus no further therapy, Outcome 2 Event‐free survival.
Figuras y tablas -
Analysis 1.2

Comparison 1 Retinoic acid versus no further therapy, Outcome 2 Event‐free survival.

Summary of findings for the main comparison. Retinoic acid postconsolidation therapy compared to no further treatment for high‐risk neuroblastoma patients treated with autologous haematopoietic stem cell transplantation

Retinoic acid postconsolidation therapy compared to no further treatment for high‐risk neuroblastoma patients treated with autologous HSCT

Patient or population: high‐risk neuroblastoma patients treated with autologous HSCT
Settings: paediatric oncology departments
Intervention: retinoic acid postconsolidation therapy
Comparison: no further treatment

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

No further treatment

Retinoic acid post‐consolidation therapy

Overall survival (reported as mortality)

583 per 10001

533 per 1000
(331 to 760)

HR 0.87
(0.46 to 1.63)

98
(1 study)

⊕⊕⊝⊝
low2,3

The length of follow‐up was not mentioned for the 98 participants eligible for this review

Treatment‐related mortality ‐ not reported

See comment

See comment

Not estimable

See comment

No adequate information on this outcome was provided

Progression‐free survival ‐ not reported

See comment

See comment

Not estimable

See comment

No information on this outcome was provided

Event‐free survival (reported as relapse, disease progression, death from any cause, or second neoplasm)

604 per 10001

549 per 1000
(371 to 749)

HR 0.86
(0.50 to 1.49)

98
(1 study)

⊕⊕⊝⊝
low2,3

The length of follow‐up was not mentioned for the 98 participants eligible for this review

Early toxicity ‐ not reported

See comment

See comment

Not estimable

See comment

No adequate information on this outcome was provided

Late toxicity including secondary malignancies ‐ not reported

See comment

See comment

Not estimable

See comment

No adequate information on this outcome was provided

Health‐related quality of life ‐ not reported

See comment

See comment

Not estimable

See comment

No information on this outcome was provided

*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% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; HR: Hazard ratio; HSCT: haematopoietic stem cell transplantation

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

1The assumed risk is based on the number of events in the control group at the final time point of the survival curve presented in the included study.
2The presence of selection bias, performance bias, attrition bias and other bias was unclear, so we downgraded one level for study limitations.
3Since this is a small study and the 95% CIs include values favouring both the intervention and the control treatment we downgraded one level for imprecision.

Figuras y tablas -
Summary of findings for the main comparison. Retinoic acid postconsolidation therapy compared to no further treatment for high‐risk neuroblastoma patients treated with autologous haematopoietic stem cell transplantation
Table 1. The INRG consensus pretreatment classification scheme

INRG stage

Age (months)

Histologic category

Grade of tumour differentiation

MYCN

11q aberration

Ploidy

Pretreatment risk group

Code

Interpretation

L1/L2

Ganglioneuroma maturing; ganglioneuroblastoma intermixed

A

Very low

L1

Any, except ganglioneuroma or ganglioneuroblastoma

Not amplified

B

Very low

Amplified

K

High

L2

< 18

Any, except ganglioneuroma or ganglioneuroblastoma

Not amplified

No

D

Low

Yes

G

Intermediate

≥ 18

Ganglioneuroblastoma nodular; neuroblastoma

Differentiating

Not amplified

No

E

Low

Yes

H

Intermediate

Poorly differentiated or undifferentiated

Not amplified

H

Intermediate

Amplified

N

High

M

< 18

Not amplified

Hyperdiploid

F

Low

< 12

Not amplified

Diploid

I

Intermediate

12 to < 18

Not amplified

Diploid

J

Intermediate

< 18

Amplified

O

High

≥ 18

P

High

MS

< 18

Not amplified

No

C

Very low

Yes

Q

High

Amplified

R

High

Reference: Cohn 2009.

The INRG consensus classification schema includes the criteria INRG stage, age, histologic category, grade of tumour differentiation, MYCN status, presence/absence of 11q aberrations, and tumour cell ploidy. Sixteen statistically or clinically different pretreatment groups of patients (lettered A through R), or both, were identified using these criteria. The categories were designated as very low (A, B, C), low (D, E, F), intermediate (G, H, I, J), or high (K, N, O, P, Q, R) pretreatment risk subsets.

Figuras y tablas -
Table 1. The INRG consensus pretreatment classification scheme
Table 2. The International Neuroblastoma Staging System

Stage

Definition

1

Localised tumour with complete gross excision, with or without microscopic residual disease; representative ipsilateral lymph nodes negative for tumour microscopically (nodes attached to and removed with the primary tumour may be positive)

2A

Localised tumour with incomplete gross excision; representative ipsilateral nonadherent lymph nodes negative for tumour microscopically

2B

Localised tumour with or without complete gross excision, with ipsilateral nonadherent lymph nodes positive for tumour. Enlarged contralateral lymph nodes must be negative microscopically

3

Unresectable unilateral tumour infiltrating across the midlinea, with or without regional lymph node involvement; or localised unilateral tumour with contralateral regional lymph node involvement; or midline tumour with bilateral extension by infiltration (unresectable) or by lymph node involvement

4

Any primary tumour with dissemination to distant lymph nodes, bone, bone marrow, liver, skin and/or other organs (except as defined for stage 4S)

4S

Localised primary tumour (as defined for stage 1, 2A or 2B), with dissemination limited to skin, liver, and/or bone marrowb (limited to infants < 1 year of age)

Reference: Brodeur 1993.

Note: Multifocal primary tumours leg, bilateral adrenal primary tumours should be staged according to the greatest extent of disease, as defined above, and followed by a subscript letter M e.g. 3M.
aThe midline is defined as the vertebral column. Tumours originating on one side and crossing the midline must infiltrate to or beyond the opposite side of the vertebral column.
bMarrow involvement in stage 4S should be minimal, i.e. < 10% of total nucleated cells identified as malignant on bone marrow biopsy or on marrow aspirate. More extensive marrow involvement would be considered to be stage 4. The (Meta‐iodobenzylguanidine) MIBG scan (if performed) should be negative in the marrow.

Figuras y tablas -
Table 2. The International Neuroblastoma Staging System
Table 3. Response to treatment

Response

Primary tumour

Metastatic sites

Complete response

No tumour

No tumour; catecholamines normal

Very good partial response

Decreased by 90% to 99%

No tumour; catecholamines normal; residual 99Tc bone changes allowed

Partial response

Decreased by more than 50%

All measurable sites decreased by > 50%. Bones and bone marrow: number of positive bone sites decreased by > 50%; no more than 1 positive bone marrow site allowed

Minimal response

No new lesions; > 50% reduction of any measurable lesion (primary or metastases) with < 50% reduction in any other; < 25% increase in any existing lesion

No response

No new lesions; < 50% reduction but < 25% increase in any existing lesion

Progressive disease

Any new lesion; increase of any measurable lesion by > 25%; previous negative marrow positive for tumour

Reference: Brodeur 1993.

Figuras y tablas -
Table 3. Response to treatment
Table 4. Children's Oncology Group assignment to low, intermediate, and high‐risk group

INSS stage

Age

MYCN

INPC classification

DNA index

Risk group

1

0 to 21 y

Any

Any

Any

Low

2A/2B

< 365 d

Any

Any

Any

Low

≥ 365 d to 21 y

Nonamplified

Any

Low

≥ 365 d to 21 y

Amplified

Favorable

Low

≥ 365 d to 21 y

Amplified

Unfavorable

High

3

< 365 d

Nonamplified

Any

Any

Intermediate

< 365 d

Amplified

Any

Any

High

≥ 365 d to 21 y

Nonamplified

Favorable

Intermediate

≥ 365 d to 21 y

Nonamplified

Unfavorable

High

≥ 365 d to 21 y

Amplified

Any

High

4

< 548 d

Nonamplified

Any

Any

Intermediate

< 365 d

Amplified

Any

Any

High

≥ 548 d to 21 y

Any

Any

High

4S

< 365 d

Nonamplified

Favorable

> 1

Low

< 365 d

Nonamplified

Any

= 1

Intermediate

< 365 d

Nonamplified

Unfavorable

Any

Intermediate

< 365 d

Amplified

Any

Any

High

Reference: NCI PDQ 2017
DNA index: favorable > 1 (hyperdiploid) or < 1 (hypodiploid); unfavorable = 1 (diploid).
Abbreviations. d: days of age; y: years of age.

Figuras y tablas -
Table 4. Children's Oncology Group assignment to low, intermediate, and high‐risk group
Table 5. Toxicity possibly associated with retinoic acid after high‐dose chemotherapy followed by autologous haematopoietic stem cell transplantation

Study

Study design

Type of HSCT

Type of RA

Dose1

Pat2

Type of adverse event (N of affected participants)

Clarke 2003

CR

PBSCT

CRA

160

1

Pneumocystis carinii pneumonia (1)

Cross 2009

CR

NR

CRA

160

1

Hypercalcaemia (1), osteoblastic lesions (1)

De Kraker 2008

SA‐IS

BMT or PBSCT

CRA

160

44

NR

Granger 2012

SA‐IS

PBSCT

CRA

160

33

NR

Hamidieh 2012

SA‐IS

PBSCT

CRA

120 to 160

14

NR

Haysom 2005

CR

BMT

CRA

160

2

Bone marrow transplant nephropathy (2)

Inamo 1999

CR

BMT

CRA

33 to 1023

1

Growth failure (1)

Khan 1996

SA‐IS

BMT

CRA

100 to 200

31

Grade 3/4 toxicity of skin, liver and hypercalcaemia correlated with peak serum levels of CRA

Kletzel 2002

SA‐IS

PBSCT

CRA

160

12

Ataxia (1)

Kogner 2004

RCT

BMT

CRA

NR

12

NR

Kohler 2000

RCT

NR

CRA

15 to 224

NR

Dry skin (47), cheilitis (24), bone pain (16), other (13)

Kreissman 2013

RCT

PBSCT

CRA

160

192

Grade 3 toxic effects: hypertension (4), hematuria (2), elevated serum creatinine (2), proteinuria (3); purged and non‐purged transplantation group combined

Kushner 2003a

CS

NR

CRA

160

1

Cheilitis (1)

Laskin 2011

CS

NR

CRA

NR

20

NR

Marabelle 2009

CR

NR

CRA

160

3

Hypercalcaemia (3)

Marmor 2008

CR

NR

Fenretinide

666 to 20515

2

Rod electroretinogram suppression (2)

Mastrangelo 2011

SA‐IS

PBSCT

CRA

160

8

NR

Matthay 2006

SA‐IS

BMT

CRA

160

22

NR

Mugishima 2008

CR

BMT

CRA

130 to 400

2

Hypercalcaemia (2)

Nishimura 1997

CR

BMT

CRA

33 to 1023

1

Generalised metaphyseal modification (1)

Park 2011

SA‐IS

PBSCT

CRA

160

30

NR

Rayburg 2009

CR

NR

Fenretinide

2210

1

Langerhans cell histiocytosis (1)

Saarinen‐Pihkala 2012

SA‐IS

BMT or PBSCT

CRA

NR

36

NR

Simon 2011

SA‐IS

NR

CRA

160

75

NR

Sirachainan 2008

CR

NR

CRA

140

1

NR

Sung 2007

SA‐IS

PBSCT

CRA

125

44

Skin eruption, particularly face

Turman 1999

CR

BMT

CRA

160

2

Bone marrow transplant nephropathy (2)

Veal 2007

SA‐IS

NR

CRA

160

28

Mild skin toxicity (9), cheilitis (1), hypercalcaemia (2)

Veal 2013

SA‐IS

NR

CRA

160

103

Grade 3 ‐ 4 skin toxicity or cheilitis (5)

Villablanca 1993

SA‐IS

BMT

CRA

100 to 200

49

Dose‐limiting toxicity of hypercalcaemia (3), arthralgia and myalgia (1), grade 1 to 3 hypercalcaemia (9)

Villablanca 1995

SA‐IS

BMT

CRA

200

51

Hypercalcaemia (3), rash (2)

Villablanca 2011

SA‐IS

NR

Fenretinide

1800 to 2475

626

Rash (1), diarrhoea (1), nausea (2), vomiting (1), nyktalopia (1), abdominal pain (4)

Yu 2010

RCT

NR

CRA

160

108

"Few toxic effects"

Notice: Studies presented in this table were not eligible for inclusion in this review.

1Dose in mg/m2/day. The unit mg/kg may be transformed to mg/m2. The average body weight, body length, and body surface of a 6‐month‐old child may be 8 kg, 67 cm, and 0.39 m2, thus 8 kg divided by 0.39 m2 is roughly equalto a conversion factor of 20 (CDC 2000a). This factor increases continuously with age. The average body weight, body length, body surface of an 11‐year‐old child may be 36 kg, 143 cm, and 1.20 m2, thus 36 kg divided by 1.20 m2 is roughly equal to a conversion factor of 30 (CDC 2000b). The unit mg per day may be transformed to m2 per day. For example, 300 mg per day may vary on average between 769 mg/m2 (300 mg/0.39 m2) and 250 m2 (300 mg/1.20 m2)

2Participants treated with retinoic acid after HDCT followed by autologous HSCT acid and evaluated for toxicity.

3Inamo 1999; Nishimura 1997: Patients received retinoic acid at a dose of 40 mg per day. The daily dose may vary on average between 102 mg/m2 (40 mg/0.39 m2) and 33 m2 (40 mg/1.20 m2)

4Kohler 2000: Participants received retinoic acid at a dose of 0.75 mg/kg per day. The daily dose may vary on average between 15 mg/m2 (0.75 mg/kg * 20) and 22 mg/m2 (0.75 mg/kg * 30).

5Marmor 2008: Participants received retinoic acid at a dose of 800 mg per day. The daily dose may vary on average between 2051 mg/m2 (800 mg/0.39 m2) and 666 m2 (800 mg/1.20 m2).

6Villablanca 2011: 51 of the 62 participants received HSCT.

BMT: bone marrow transplantation; CR: case report; CS: case series; CRA: 13‐cis‐retinoic acid; HDCT: high‐dose chemotherapy; HSCT: haematopoietic stem cell transplantation; N: number of participants; NR: not reported; PBSCT: peripheral blood stem cell transplantation; RA: retinoic acid; SA‐IS: single‐arm intervention study such as phase‐1 or phase‐2 clinical trial

Figuras y tablas -
Table 5. Toxicity possibly associated with retinoic acid after high‐dose chemotherapy followed by autologous haematopoietic stem cell transplantation
Comparison 1. Retinoic acid versus no further therapy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.87 [0.46, 1.63]

2 Event‐free survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

0.86 [0.50, 1.49]

Figuras y tablas -
Comparison 1. Retinoic acid versus no further therapy