Scolaris Content Display Scolaris Content Display

Interventions for preventing oral mucositis in patients with cancer receiving treatment: cytokines and growth factors

Contraer todo Desplegar todo

Referencias

Antoun 2009 {published data only}

Antoun S, Boige V, Ducreux M, Paintain M, van't Hof M, Enslen M, et al. Protective effect of an enteral formula containing TGF‐beta(2) in the prevention of chemotherapy‐induced diarrhoea: a pilot study. e‐SPEN, European e‐Journal of Clinical Nutrition and Metabolism 2009;4(6):e348‐50. CENTRAL

Blazar 2006 {published data only}

Blazar BR, Weisdorf DJ, Defor T, Goldman A, Braun T, Silver S, et al. Phase 1/2 randomized, placebo‐control trial of palifermin to prevent graft‐versus‐host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (HSCT). Blood 2006;108(9):3216‐22. CENTRAL

Blijlevens 2013 {published data only}

Blijlevens N, de Château M, Krivan G, Rabitsch W, Szomor A, Pytik R, et al. In a high‐dose melphalan setting, palifermin compared to placebo had no effect on oral mucositis or related patient's burden. Supportive Care in Cancer 2011;19(Suppl 2):S227‐8. CENTRAL
Blijlevens N, de Château M, Krivan G, Rabitsch W, Szomor A, Pytik R, et al. Medical resource use of two different dosing regimens of palifermin to prevent mucositis in multiple myeloma patients receiving one‐day administration of high‐dose melphalan. Bone Marrow Transplantation 2011;46(Suppl 1s):S163‐4. CENTRAL
Blijlevens N, de Château M, Krivan G, Rabitsch W, Szomor A, Pytik R, et al. Randomized controlled trial of two different dosing regimens of palifermin to prevent mucositis in multiple myeloma patients receiving one‐day administration of high‐dose melphalan. Blood 2010;116(21):904. CENTRAL
Blijlevens N, de Château M, Krivan G, Rabitsch W, Szomor A, Pytlik R, et al. In a high‐dose melphalan setting, palifermin compared with placebo had no effect on oral mucositis or related patient's burden. Bone Marrow Transplantation 2013;48(7):966‐71. CENTRAL

Bradstock 2014 {published data only}

Bradstock KF, Link E, Collins M, Di Iulio J, Lewis ID, Schwarer A, et al. A randomized trial of prophylactic palifermin on gastrointestinal toxicity after intensive induction therapy for acute myeloid leukaemia. British Journal of Haematology 2014;167(5):618‐25. CENTRAL

Brizel 2008 {published data only}

Brizel DM, Murphy BA, Rosenthal DI, Pandya KJ, Glück S, Brizel HE, et al. Phase II study of palifermin and concurrent chemoradiation in head and neck squamous cell carcinoma. Journal of Clinical Oncology 2008;26(15):2489‐96. CENTRAL

Cartee 1995 {published data only}

Cartee L, Petros WP, Rosner G, Gilbert C, Moore S, Affronti M, et al. Evaluation of GM‐CSF mouthwash for prevention of chemotherapy‐induced mucositis: a randomized double‐blind dose escalating study. Blood 1993;82(10 Suppl 1):496a. [Abs No 1968]CENTRAL
Cartee L, Petros WP, Rosner GL, Gilbert C, Moore S, Affronti ML, et al. Evaluation of GM‐CSF mouthwash for prevention of chemotherapy‐induced mucositis: a randomized, double‐blind, dose‐ranging study. Cytokine 1995;7(5):471‐7. CENTRAL

Cesaro 2013 {published data only}

Cesaro S, Nesi F, Tridello G, Abate M, Panizzolo IS, Balter R, et al. A randomized, non‐inferiority study comparing efficacy and safety of a single dose of pegfilgrastim versus daily filgrastim in pediatric patients after autologous peripheral blood stem cell transplant. PLOS One 2013;8(1):e53252. CENTRAL
Cesaro S, Nesi F, Tridello G, Fagioli F, Abate M, Panizzolo I, et al. A non‐inferiority study comparing efficacy and safety of a single dose of pegfilgrastim versus daily filgrastim in paediatric patients after autologous peripheral blood stem cell transplant. Bone Marrow Transplantation 2012;47(Suppl 1s):S372. CENTRAL

Chi 1995 {published data only}

Chi KH, Chen CH, Chan WK, Chow KC, Chen SY, Yen SH, et al. Effect of granulocyte‐macrophage colony‐stimulating factor on oral mucositis in head and neck cancer patients after cisplatin, fluorouracil and leucovorin chemotherapy. Journal of Clinical Oncology 1995;13(10):2620‐8. CENTRAL
Chi KH, Chen CH, Chan WK, Yen SH, Liang MJ, Chou KC, et al. Effect of granulocyte‐macrophage colony‐stimulating factor (GH‐CFS) on oral mucositis in head and neck cancer patients after cisplatin, 5‐FU and leucovorin chemotherapy. Proceedings of the American Society of Clinical Oncology 1994;13:428. [Abs No 1469]CENTRAL

Crawford 1999 {published data only}

Crawford J, Glaspy J, Vincent M, Tomita D, Mazanet R. Effect of filgrastim (r‐metHuG‐CSF) on oral mucositis in patients with small cell lung cancer (SCLC) receiving chemotherapy (cyclophosphamide, doxorubicin and etoposide, CAE). Proceedings of the American Society of Clinical Oncology 1994;13:442. [Abs No A1523]CENTRAL
Crawford J, Ozer H, Stoller R, Johnson D, Lyman G, Tabbara I, et al. Reduction by granulocyte colony‐stimulating factor of fever and neutropenia induced by chemotherapy in patients with small‐cell lung cancer. New England Journal of Medicine 1991;325(3):164‐70. CENTRAL
Crawford J, Tomita DK, Mazanet R, Glaspy J, Ozer H. Reduction of oral mucositis by filgrastim (r‐metHuG‐CSF) in patients receiving chemotherapy. Cytokines, Cellular and Molecular Therapy 1999;5(4):187‐93. CENTRAL

Dazzi 2003 {published data only}

Dazzi C, Cariello A, Giovanis P, Monti M, Vertogen B, Leoni M, et al. Prophylaxis with GM‐CSF mouthwashes does not reduce frequency and duration of severe oral mucositis in patients with solid tumors undergoing high‐dose chemotherapy with autologous peripheral blood stem cell transplantation rescue: a double‐blind, randomized, placebo‐controlled study. Annals of Oncology 2003;14(4):559‐63. CENTRAL
Dazzi C, Cariello A, Monti M, Giovanis P, Vertogen B, Nanni O, et al. Prophylaxis with GM‐CSF mouthwash does not reduce frequency and duration of severe oral mucositis in patients with solid tumors undergoing high dose chemotherapy with autologous PBPC rescue: a double‐blind randomized placebo‐controlled study. Annals of Oncology 2002;13 Suppl 5:167. [Abs No 617O]CENTRAL

Fink 2011 {published and unpublished data}

Fink G, Ihorst G, Burbeck M, Kleber M, Kohlweyer U, Engelhardt M. Prospective randomized trial of palifermin (keratinocyte growth factor) versus best supportive care measures (BSC) for the decrease of oral mucositis (OM) in lymphoma patients receiving high‐dose BEAM‐(BCNU, Etoposide, Ara‐C, Melphalan) conditioning and autologous stem‐cell transplantation (ASCT). Onkologie. Jahrestagung der Deutschen, Österreichischen und Schweizerischen Gesellschaften für Hämatologie und Onkologie; 2011 Sept‐Oct 30‐04; Basel (Switzerland). Basel (Switzerland): S Karger AG, 2011:302. CENTRAL

Freytes 2004 {published data only}

Freytes CO, Ratanatharathorn V, Taylor C, Abboud C, Chesser N, Restrepo A, et al. Phase I/II randomized trial evaluating the safety and clinical effects of repifermin administered to reduce mucositis in patients undergoing autologous hematopoietic stem cell transplantation. Clinical Cancer Research 2004;10(24):8318‐24. CENTRAL

Gholizadeh 2016 {published data only}

Gholizadeh N, Mehdipoor M, Sajadi H, Moosavi MS. Palifermin and chlorhexidine mouthwashes in prevention of chemotherapy‐induced mucositis in children with acute lymphocytic leukemia: a randomized controlled trial. Journal of Dentistry (Shiraz, Iran) 2016;17(4):343‐7. CENTRAL

Henke 2011 {published data only}

Henke M, Alfonsi M, Foa P, Giralt J, Bardet E, Cerezo L, et al. Palifermin decreases severe oral mucositis of patients undergoing postoperative radiochemotherapy for head and neck cancer: a randomized, placebo‐controlled trial. Journal of Clinical Oncology 2011;29(20):2815‐20. CENTRAL
Henke M, Alfonsi M, Foa P, Giralt J, Bardet E, Cerezo L, et al. Palifermin significantly reduces severe oral mucositis in subjects with resected locally advanced head and neck cancer undergoing post‐operative concurrent radio‐chemotherapy. Radiotherapy and Oncology 2008;88 Suppl 2:S152. CENTRAL

Hosseinjani 2017 {published data only}

Hosseinjani H, Hadjibabaie M, Gholami K, Javadi M, Radfar M, Jahangard‐Rafsanjani Z, et al. The efficacy of erythropoietin mouthwash in prevention of oral mucositis in patients undergoing autologous hematopoietic SCT: a double‐blind, randomized, placebo‐controlled trial. Hematological Oncology 2017;35(1):106‐12. CENTRAL

Jagasia 2012 {published data only}

Jagasia MH, Abonour R, Long GD, Bolwell BJ, Laport GG, Shore TB, et al. Palifermin for the reduction of acute GVHD: a randomized, double‐blind, placebo‐controlled trial. Bone Marrow Transplantation 2012;47(10):1350‐5. CENTRAL

Katano 1995 {published data only}

Katano M, Nakamura M, Matsuo T, Iyama A, Hisatsugu T. Effect of granulocyte colony‐stimulating factor (G‐CSF) on chemotherapy‐induced oral mucositis. Surgery Today 1995;25(3):202‐6. CENTRAL

Kim 2017 {published data only}

Kim J‐W, Kim KI, Lee HJ, Kim B‐S, Bang S‐M, Kim I, et al. Interim analysis of randomized phase II study of recombinant human epidermal growth factor on oral mucositis induced by intensive chemotherapy with stem cell transplantation for hematologic malignancies. Blood 2010;116(21):905. CENTRAL
Kim JW, Kim MG, Lee HJ, Koh Y, Kwon JH, Kim I, et al. Topical recombinant human epidermal growth factor for oral mucositis induced by intensive chemotherapy with hematopoietic stem cell transplantation: final analysis of a randomized, double‐blind, placebo‐controlled, phase 2 trial. PLOS One 2017;12(1):e0168854. CENTRAL
Kim KI, Kim JW, Lee HJ, Kim BS, Bang SM, Kim I, et al. Recombinant human epidermal growth factor on oral mucositis induced by intensive chemotherapy with stem cell transplantation. American Journal of Hematology 2013;88(2):107‐12. CENTRAL

Le 2011 {published data only}

Le Q, Kim H, Schneider C, Muraközy G, Skladowski K, Reinisch S, et al. Palifermin reduces severe oral mucositis in subjects with locally advanced head and neck cancer undergoing chemoradiotherapy. International Journal of Radiation Oncology, Biology, Physics 2008;72 Suppl 1:S32. CENTRAL
Le QT, Kim HE, Schneider CJ, Muraközy G, Skladowski K, Reinisch S, et al. Palifermin reduces severe mucositis in definitive chemoradiotherapy of locally advanced head and neck cancer: a randomized, placebo‐controlled study. Journal of Clinical Oncology 2011;29(20):2808‐14. CENTRAL

Linch 1993 {published data only}

Linch DC, Scarffe H, Proctor S, Chopra R, Taylor PR, Morgenstern G, et al. Randomised vehicle‐controlled dose‐finding study of glycosylated recombinant human granulocyte colony‐stimulating factor after bone marrow transplantation. Bone Marrow Transplantation 1993;11(4):307‐11. CENTRAL

Lucchese 2016a {published data only}

Lucchese A, Matarese G, Ghislanzoni LH, Gastaldi G, Manuelli M, Gherlone E. Efficacy and effects of palifermin for the treatment of oral mucositis in patients affected by acute lymphoblastic leukemia. Leukemia and Lymphoma 2016;57(4):820‐7. CENTRAL

Lucchese 2016b {published data only}

Lucchese A, Matarese G, Manuelli M, Ciuffreda C, Bassani L, Isola G, et al. Reliability and efficacy of palifermin in prevention and management of oral mucositis in patients with acute lymphoblastic leukemia: a randomized, double‐blind controlled clinical trial. Minerva Stomatologica 2016;65(1):43‐53. CENTRAL

Makkonen 2000 {published data only}

Makkonen TA, Minn H, Jekunen A, Vilja P, Tuominen J, Joensuu H. Granulocyte macrophage‐colony stimulating factor (GM‐CSF) and sucralfate in prevention of radiation‐induced mucositis: a prospective randomized study. International Journal of Radiation Oncology, Biology, Physics 2000;46(3):525‐34. CENTRAL
Minn HR, Makkonen TA, Jekunen A, Vilja P, Tuominen J, Joensuu H. Granulocyte macrophage‐colony stimulating factor (GM‐CSF) and sucralfate in prevention of radiation‐induced mucositis: a prospective randomized study. International Journal of Radiation Oncology, Biology, Physics 1999;45 Suppl 3:239. CENTRAL

McAleese 2006 {published data only}

McAleese JJ, Bishop KM, A'Hern R, Henk JM. Randomized phase II study of GM‐CSF to reduce mucositis caused by accelerated radiotherapy of laryngeal cancer. British Journal of Radiology 2006;79(943):608‐13. CENTRAL

Meropol 2003 {published data only}

Meropol NJ, Somer RA, Gutheil J, Pelley RJ, Modiano MR, Rowinsky EK, et al. Randomized phase I trial of recombinant human keratinocyte growth factor plus chemotherapy: potential role as mucosal protectant. Journal of Clinical Oncology 2003;21(8):1452‐8. CENTRAL

Nemunaitis 1995 {published data only}

Nemunaitis J, Rosenfeld CS, Ash R, Freedman MH, Deeg HJ, Appelbaum F, et al. Phase III randomized, double‐blind placebo‐controlled trial of rhGM‐CSF following allogeneic bone marrow transplantation. Bone Marrow Transplantation 1995;15(6):949‐54. CENTRAL

Peterson 2009 {published data only}

Barker NP, Peterson DE, Akhmadullina LI, Rodionova I, Sherman NZ, Gertner JM, et al. Prophylaxis of recurrent chemotherapy‐induced oral mucositis: a phase II multicenter, randomized, placebo‐controlled trial of recombinant human intestinal trefoil factor (rhITF). Journal of Clinical Oncology 2008;26:505. [Abs No 9514]CENTRAL
Peterson DE, Barker NP, Akhmadullina LI, Rodionova I, Sherman NZ, Davidenko IS, et al. Phase II, randomized, double‐blind, placebo‐controlled study of recombinant human intestinal trefoil factor oral spray for prevention of oral mucositis in patients with colorectal cancer who are receiving fluorouracil‐based chemotherapy. Journal of Clinical Oncology 2009;27(26):4333‐8. CENTRAL

Rosen 2006 {published data only}

Clarke SJ, Abdi E, Davis ID, Schnell FM, Zalcberg JR, Gutheil J, et al. Recombinant human keratinocyte growth factor (rHuKGF) prevents chemotherapy‐induced mucositis in patients with advanced colorectal cancer: a randomized phase II trial. Proceedings of the American Society of Clinical Oncology 2001;20 (Pt 1):383a. [Abs No 1529]CENTRAL
Rosen LS, Abdi E, Davis ID, Gutheil J, Schnell FM, Zalcberg J, et al. Palifermin reduces the incidence of oral mucositis in patients with metastatic colorectal cancer treated with fluorouracil‐based chemotherapy. Journal of Clinical Oncology 2006;24(33):5194‐200. CENTRAL

Saarilahti 2002 {published data only}

Saarilahti K, Kajanti M, Joensuu T, Kouri M, Joensuu H. Comparison of granulocyte‐macrophage colony‐stimulating factor and sucralfate mouthwashes in the prevention of radiation‐induced mucositis: a double‐blind prospective randomized phase III study. International Journal of Radiation Oncology, Biology, Physics 2002;54(2):479‐85. CENTRAL

Schneider 1999 {published data only}

Schneider SB, Nishimura RD, Zimmerman RP, Tran L, Shiplacoff J, Tormey M, et al. Filgrastim (r‐metHuG‐CSF) and its potential use in the reduction of radiation‐induced oropharyngeal mucositis: an interim look at a randomized, double‐blind, placebo‐controlled trial. Cytokines, Cellular and Molecular Therapy 1999;5(3):175‐80. CENTRAL

Spielberger 2004 {published data only}

Emmanouilides C, Spielberger R, Stiff P, Rong A, Isitt J, Bensinger W, et al. Palifermin treatment of mucositis in transplant patients reduces health resource use: phase III results. Journal of Supportive Oncology 2004;2(2):77‐8. CENTRAL
Spielberger R, Emmanouilides C, Stiff P, Bensinger W, Gentile T, Weisdorf D, et al. Use of recombinant human keratinocyte growth factor (palifermin) can reduce severe oral mucositis in patients with hematologic malignancies undergoing autologous peripheral blood progenitor cell transplantation after radiation‐based conditioning. Journal of Supportive Oncology 2004;2(2):73‐4. CENTRAL
Spielberger R, Stiff P, Bensinger W, Gentile T, Weisdorf D, Kewalramani T, et al. Palifermin for oral mucositis after intensive therapy for hematologic cancers. New England Journal of Medicine 2004;351(25):2590‐8. CENTRAL
Stiff P, Bensinger W, Emmanouilides C, Gentile T, Weisdorf D, Shea T, et al. Treatment of mucositis with palifermin improves patient function and results in a clinically meaningful reduction in mouth and throat soreness: phase III results. Journal of Supportive Oncology 2004;2(2):75‐6. CENTRAL
Stiff PJ, Emmanouilides C, Bensinger WI, Gentile T, Blazar B, Shea TC, et al. Palifermin reduces patient‐reported mouth and throat soreness and improves patient functioning in the hematopoietic stem‐cell transplantation setting. Journal of Clinical Oncology 2006;24(33):5186‐93. CENTRAL

Su 2006 {published data only}

Su YB. Double‐blind, randomized trial of granulocyte‐colony stimulating factor (GCSF) versus (v) placebo during postoperative radiation (RT) for advanced resectable squamous cell head and neck cancer (SCCHN): impact on mucositis. Journal of Clinical Oncology 2004;22 Suppl:14S. CENTRAL
Su YB, Vickers AJ, Zelefsky MJ, Kraus DH, Shaha AR, Shah JP, et al. Double‐blind, placebo‐controlled, randomized trial of granulocyte‐colony stimulating factor during postoperative radiotherapy for squamous head and neck cancer. Cancer Journal 2006;12(3):182‐8. CENTRAL

Vadhan‐Raj 2010 {published data only}

Vadhan‐Raj S, Trent J, Patel S, Zhou X, Johnson MM, Araujo D, et al. Single‐dose palifermin prevents severe oral mucositis during multicycle chemotherapy in patients with cancer: a randomised trial. Annals of Internal Medicine 2010;153(6):358‐67. CENTRAL
Vadhan‐Raj S, Trent JC, Patel SR, Araujo DM, Ludwig LA, Bailey D, et al. Randomized, double‐blind, placebo‐controlled study of palifermin for the prevention of mucositis in patients receiving doxorubicin‐based chemotherapy. Journal of Clinical Oncology 2008;26(15 Suppl):9547. CENTRAL

van der Lelie 2001 {published data only}

van der Lelie H, Thomas BL, van Oers RH, Ek‐Post M, Sjamsoedin SA, van Dijk‐Overtoom ML, et al. Effect of locally applied GM‐CSF on oral mucositis after stem cell transplantation: a prospective placebo‐controlled double‐blind study. Annals of Hematology 2001;80(3):150‐4. CENTRAL

Wu 2009 {published data only}

Ahn Y, Wu H, Song S, Kim Y, Oh Y, Lee C, et al. The therapeutic effect of recombinant human epidermal growth factor (rhEGF) on mucositis in patients with head and neck cancer undergoing radiotherapy with or without chemotherapy: a double‐blind placebo‐controlled prospective phase II multi‐institutional study. Journal of Clinical Oncology 2008;26(15 Suppl):6021. CENTRAL
Lee S, Song S, Kim Y, Oh Y, Lee C, Keum K, et al. The therapeutic effect of recombinant human epidermal growth factor (rhEGF) on mucositis in patients with head and neck cancer undergoing radiotherapy with or without chemotherapy: a double‐blind placebo‐controlled prospective phase II multi‐institutional clinical trial. International Journal of Radiation Oncology, Biology, Physics 2008;72(1):S32. CENTRAL
Wu HG, Song SY, Kim YS, Oh YT, Lee CG, Keum KC, et al. Therapeutic effect of recombinant human epidermal growth factor (RhEGF) on mucositis in patients undergoing radiotherapy, with or without chemotherapy, for head and neck cancer: a double‐blind placebo‐controlled prospective phase 2 multi‐institutional clinical trial. Cancer 2009;115(16):3699‐708. CENTRAL

Antin 2002 {published data only}

Antin JH, Lee SJ, Neuberg D, Alyea E, Soiffer RJ, Sonis S, et al. A phase I/II double‐blind, placebo‐controlled study of recombinant human interleukin‐11 for mucositis and acute GVHD prevention in allogeneic stem cell transplantation. Bone Marrow Transplantation 2002;29(5):373‐7. CENTRAL

de Koning 2007 {published data only}

de Koning BA, Philipsen‐Geerling B, Hoijer M, Hahlen K, Buller HA, Pieters R. Protection against chemotherapy induced mucositis by TGF‐beta(2) in childhood cancer patients: results from a randomized cross‐over study. Pediatric Blood & Cancer 2007;48(5):532‐9. CENTRAL

Foncuberta 2001 {published data only}

Foncuberta MC, Cagnoni PJ, Brandts CH, Mandanas R, Fields K, Derigs HG, et al. Topical transforming growth factor‐beta3 in the prevention or alleviation of chemotherapy‐induced oral mucositis in patients with lymphomas or solid tumors. Journal of Immunotherapy 2001;24(4):384‐8. CENTRAL

Gebbia 1994 {published data only}

Gebbia V, Valenza R, Testa A, Cannata G, Borsellino N, Gebbia N. A prospective randomized trial of thymopentin versus granulocyte‐colony stimulating factor with or without thymopentin in the prevention of febrile episodes in cancer patients undergoing highly cytotoxic chemotherapy. Anticancer Research 1994;14(2B):731‐4. CENTRAL

Gladkov 2013 {published data only}

Gladkov O, Buchner A, Bias P, Mueller U, Elsaesser R. Chemotherapy‐associated treatment burden in breast cancer patients receiving lipegfilgrastim or pegfilgrastim: secondary efficacy data from a phase III study. Haematologica 2013;98(Suppl 1):426. CENTRAL

Gordon 1993 {published data only}

Gordon B, Spadinger A, Hodges E, Coccia P. Effect of granulocyte macrophage colony stimulating factor (GMCSF) on oral mucositis after autologous bone marrow transplantation. Proceedings of the American Society of Clinical Oncology 1993;12:432. [Abs No 1489]CENTRAL

Horsley 2007 {published data only}

Horsley P, Bauer JD, Mazkowiack R, Gardner R, Bashford J. Palifermin improves severe mucositis, swallowing problems, nutrition impact symptoms, and length of stay in patients undergoing hematopoietic stem cell transplantation. Supportive Care in Cancer 2007;15(1):105‐9. CENTRAL

Hunter 2009 {published data only}

Hunter A, Mahendra P, Wilson K, Fields P, Cook G, Peniker A, et al. A randomized, double‐blind, placebo‐controlled, multicenter trial of ATL‐104, a swallowable mouthwash, in patients with oral mucositis following peripheral blood stem cell transplantation. Journal of Supportive Oncology 2007;5(4 Suppl 2):52‐3. CENTRAL
Hunter A, Mahendra P, Wilson K, Fields P, Cook G, Peniket A, et al. Treatment of oral mucositis after peripheral blood SCT with ATL‐104 mouthwash: results from a randomized, double‐blind, placebo‐controlled trial. Bone Marrow Transplantation 2009;43(7):563‐9. CENTRAL

Ifrah 1999 {published data only}

Ifrah N, Witz F, Jouet JP, Francois S, Lamy T, Linassier C, et al. Intensive short term therapy with granulocyte‐macrophage‐colony stimulating factor support, similar to therapy for acute myeloblastic leukemia, does not improve overall results for adults with acute lymphoblastic leukemia. American Cancer Society 1999;86(8):1496‐505. CENTRAL

Iwase 1997 {published data only}

Iwase M, Yoshiya M, Kakuta S, Nagumo M. Clinical trial of recombinant granulocyte colony‐stimulating factor for chemotherapy‐induced neutropenia patients with oral cancer. Journal of Oral and Maxillofacial Surgery 1997;55(8):836‐40. CENTRAL

Jones 1996 {published data only}

Jones SE, Schottstaedt MW, Duncan LA, Kirby RL, Good RH, Mennel RG, et al. Randomized double‐blind prospective trial to evaluate the effects of sargramostim versus placebo in a moderate‐dose fluorouracil, doxorubicin, and cyclophosphamide adjuvant chemotherapy program for stage II and III breast cancer. Journal of Clinical Oncology 1996;14(11):2976‐83. CENTRAL

Karthaus 1998 {published data only}

Karthaus M, Rosenthal C, Huebner G, Paul H, Elser C, Hertenstein B, et al. Effect of topical oral G‐CSF on oral mucositis: a randomised placebo‐controlled trial. Bone Marrow Transplantation 1998;22(8):781‐5. CENTRAL
Karthaus M, Rosenthal C, Paul H, Novotny J, Hóbner G, Fenchel K, et al. Effect of topical oral G‐CSF application on oral mucositis in high‐grade lymphoma patients treated with hd‐methotrexate (hd‐mtx) ‐ results of a randomized placebo‐controlled trial. Proceedings of the American Society of Clinical Oncology 1998;17:235. CENTRAL

Kubo 2016 {published data only}

Kubo K, Miyazaki Y, Murayama T, Shimazaki R, Usui N, Urabe A, et al. A randomized, double‐blind trial of pegfilgrastim versus filgrastim for the management of neutropenia during CHASE(R) chemotherapy for malignant lymphoma. British Journal of Haematology 2016;174(4):563‐70. CENTRAL

Lee 2016 {published data only}

Lee KH, Kim JY, Lee MH, Han HS, Lim JH, Park KU, et al. A randomized, multicenter, phase II/III study to determine the optimal dose and to evaluate the efficacy and safety of pegteograstim (GCPGC) on chemotherapy‐induced neutropenia compared to pegfilgrastim in breast cancer patients: KCSG PC10‐09. Supportive Care in Cancer 2016;24(4):1709‐17. CENTRAL

Legros 1997 {published data only}

Legros M, Fleury J, Bay JO, Choufi B, Basile M, Condat P, et al. rhGM‐CSF vs placebo following rhGM‐CSF‐mobilized PBPC transplantation: a phase III double‐blind randomized trial. Bone Marrow Transplantation 1997;19(3):209‐13. CENTRAL

Limaye 2013 {published data only}

Limaye SA, Haddad RI, Cilli F, Sonis ST, Colevas AD, Brennan MT, et al. Phase 1b, multicenter, single blinded, placebo‐controlled, sequential dose escalation study to assess the safety and tolerability of topically applied AG013 in subjects with locally advanced head and neck cancer receiving induction chemotherapy. Cancer 2013;119(24):4268‐76. CENTRAL
Murphy B, Limaye SA, Sonis ST, Cilli F, Brennan MT, Hu KS, et al. Phase 1B study assessing safety, tolerability and efficacy of AG013 in subjects with head and neck cancer receiving induction chemotherapy. Supportive Care in Cancer 2012;20(1 Suppl):S259. [Abs No 1078]CENTRAL

Nabholtz 2002 {published data only}

Nabholtz JM, Cantin J, Chang J, Guevin R, Patel R, Tkaczuk K, et al. Phase III trial comparing granulocyte colony‐stimulating factor to leridistim in the prevention of neutropenic complications in breast cancer patients treated with docetaxel/doxorubicin/cyclophosphamide: results of the BCIRG 004 trial. Clinical Breast Cancer 2002;3(4):268‐75. CENTRAL

NCT00360971 {published data only}

NCT00360971. Palifermin in lessening oral mucositis in patients undergoing radiation therapy and chemotherapy for locally advanced head and neck cancer [A randomized, phase III, double‐blind, placebo‐controlled study to evaluate the efficacy and safety of palifermin (NSC# 740548; IND # 6370) for the reduction of oral mucositis in patients with locally advanced head and neck cancer receiving radiation therapy with concurrent chemotherapy (followed by surgery for selected patients)]. clinicaltrials.gov/show/NCT00360971 (first received 3 August 2006). CENTRAL

NCT00626639 {published data only}

NCT00626639. A study of palifermin for the reduction of oral mucositis in patients with locally advanced head and neck cancer receiving postoperative radiotherapy and concurrent chemotherapy [A phase 1/2 study to evaluate safety, pharmacokinetics, pharmacodynamics and preliminary efficacy of weekly doses of palifermin (rHuKGF) for the reduction of oral mucositis in subjects with locally advanced head and neck cancer (HNC) receiving postoperative radiotherapy with concurrent chemotherapy]. clinicaltrials.gov/show/NCT00626639 (first received 21 February 2008). CENTRAL

Pettengell 1992 {published data only}

Pettengell R, Gurney H, Radford JA, Deakin DP, James R, Wilkinson PM, et al. Granulocyte colony‐stimulating factor to prevent dose‐limiting neutropenia in non‐Hodgkin's lymphoma: a randomized controlled trial. Blood 1992;80(6):1430‐6. CENTRAL

Ryu 2007 {published data only}

Hoffman KE, Pugh SL, James JL, Scarantino C, Movsas B, Valicenti RK, et al. The impact of concurrent granulocyte‐macrophage colony‐stimulating factor on quality of life in head and neck cancer patients: results of the randomized, placebo‐controlled Radiation Therapy Oncology Group 9901 trial. Quality of Life Research 2014;23(6):1841‐58. CENTRAL
Ryu JK, Swann S, LeVeque F, Scarantino CW, Johnson D, Chen A, et al. The impact of concurrent granulocyte macrophage‐colony stimulating factor on radiation‐induced mucositis in head and neck cancer patients: a double‐blind placebo‐controlled prospective phase III study by Radiation Therapy Oncology Group 9901. International Journal of Radiation Oncology, Biology, Physics 2007;67(3):643‐50. CENTRAL

Throuvalas 1995 {published data only}

Throuvalas N, Antonadou D, Pulizzi M, Sarris G. Evaluation of the efficacy and safety of GM‐CSF in the prophylaxis of mucositis in patients with head and neck cancer treated with RT. European Journal of Cancer 1995;31 Suppl 5:S93. [Abs No 431]CENTRAL

Tsurusawa 2016 {published data only}

Tsurusawa M, Watanabe T, Gosho M, Mori T, Mitsui T, Sunami S, et al. Randomized study of granulocyte colony stimulating factor for childhood B‐cell non‐Hodgkin lymphoma: a report from the Japanese pediatric leukemia/lymphoma study group B‐NHL03 study. Leukemia & Lymphoma 2016;57(7):1657‐64. CENTRAL

Vitale 2014 {published data only}

Vitale KM, Violago L, Cofnas P, Bishop J, Jin Z, Bhatia M, et al. Impact of palifermin on incidence of oral mucositis and healthcare utilization in children undergoing autologous hematopoietic stem cell transplantation for malignant diseases. Pediatric Transplantation 2014;18(2):211‐6. CENTRAL

References to studies awaiting assessment

ACTRN12606000083594 {published data only}

ACTRN12606000083594. Safety and efficacy trial of whey growth factor extract for oral mucositis [A multicentre, double‐blind, placebo‐controlled, safety and efficacy trial of whey growth factor extract (WGFEA) for oral mucositis in lymphoma patients undergoing carmustine, etoposide, cytosine arabinoside and melphalan (BEAM) chemotherapy and autologous stem cell transplantation]. www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=1142 (first received 27 February 2006). CENTRAL

Antonadou 1998 {published data only}

Antonadou D, Athanassiou E, Synodinou M, Koliarakis N, Panoussaki K, Karageorgis P, et al. Evaluation of the efficacy of granulocyte macrophage colony stimulating factor (GM‐CSF) in the prevention of radiation induced mucositis. Radiotherapy and Oncology 1998;48 Suppl 1:S39. CENTRAL

Elsaid 2001 {published data only}

Elsaid A, Farouk M. Significance of anemia and role of erythropoietin in radiation induced mucositis in head and neck cancer patients. International Journal of Radiation Oncology, Biology, Physics 2001;51(3 Suppl 1):368. CENTRAL

Grzegorczyk 2006 {published data only}

Grzegorczyk‐Jazwinska A, Dwilewicz‐Trojaczek J, Kozak I, Karakulska‐Prystupiuk E, Gorska R. Effect of locally applied G‐CSF on oral mucositis after autogeneic and allogeneic stem cell transplantation. Acta Haematologica Polonica 2006;37(2):225‐40. CENTRAL
Grzegorczyk‐Jazwinska A, Dwilewicz‐Trojaczek J, Kozak I, Karakulska‐Prystupiuk E, Gorska R. Effect of locally applied G‐CSF on oral mucositis after autologic stem cell transplantation. Dental and Medical Problems 2004;41(4):695‐701. CENTRAL

Koga 2015 {published data only}

Koga Y, Tsurusawa M, Watanabe T, Gosho M, Mori T, Mitsui T, et al. Randomized study of granulocyte colony stimulating factor for childhood B‐cell non‐Hodgkin lymphoma: a report from the Japanese pediatric leukemia/lymphoma study Group B‐NHL03 study. British Journal of Haematology 2015;171(Suppl S1):63‐4. CENTRAL

NCT00293462 {published data only}

NCT00293462. GM‐CSF mouthwash for preventing and treating mucositis in patients who are undergoing radiation therapy for head and neck cancer [Management of mucositis with GM‐CSF (sargramostim) mouthwash study protocol]. clinicaltrials.gov/show/NCT00293462 (first received 16 February 2006). CENTRAL

NCT00393822 {published data only}

NCT00393822. A study of palifermin for the reduction of oral mucositis in subjects with stage 2B or 3 locally advanced, colon cancer [A randomized, double‐blind, placebo‐controlled study evaluating the efficacy and safety of palifermin (recombinant human keratinocyte growth factor) for reduction of oral mucositis in subjects with stage 2B or 3 locally advanced, colon cancer receiving 5‐FU and leucovorin as adjuvant therapy]. clinicaltrials.gov/show/NCT00393822 (first received 26 October 2006). CENTRAL

NCT02303197 {published data only}

NCT02303197. Clinical trial on the efficacy and safety of Chining decoction in the treatment of radiation stomatitis [A randomized, controlled clinical trial on the efficacy and safety of Chining decoction in the treatment of radiation stomatitis]. clinicaltrials.gov/show/NCT02303197 (first received 25 November 2014). CENTRAL

NCT02313792 {published data only}

NCT02313792. Palifermin for patients receiving hematopoietic stem cell transplantation [Efficacy, safety and quality of life of palifermin on reducing oral mucositis in patients with hematopoietic stem cell transplantation, prospective double‐blind randomized phase III trial]. clinicaltrials.gov/show/NCT02313792 (first received 5 December 2014). CENTRAL

Patte 2002 {published data only}

Patte C, Laplanche A, Bertozzi AI, Baruchel A, Frappaz D, Schmitt C, et al. Granulocyte colony‐stimulating factor in induction treatment of children with non‐Hodgkin's lymphoma: a randomized study of the French Society of Pediatric Oncology. Journal of Clinical Oncology 2002;20(2):441‐8. CENTRAL

Schuster 2007a {published data only}

Schuster MW, Anaissie E, Hurd D, Bensinger W, Mason J, McCarty J, et al. Final analysis of the phase II, randomized, double‐blind, placebo‐controlled trial of single‐dose velafermin (CG53135‐05) for the prevention of oral mucositis. Journal of Supportive Oncology 2007;5(4 Suppl 2):58‐9. CENTRAL

Schuster 2007b {published data only}

Schuster MW, Mehta J, Waller EK, Rifkin RM, Micallef I, Hurd D, et al. A randomized placebo controlled phase II trial of prevention of severe oral mucositis using single dose velafermin in patients receiving myeloablative therapy and autologous hematopoietic stem cell transplant (AHSCT). Blood 2007;110(11):615. CENTRAL

Shea 2007 {published data only}

Shea TC, Kewalramani T, Mun Y, Jayne G, Dreiling LK. Evaluation of single‐dose palifermin to reduce oral mucositis (OM) in fractionated total body irradiation (fTBI) and high dose (HD) chemotherapy with autologous peripheral blood progenitor cell (PBPC) transplantation. Blood 2006;108(11 Part 1):875‐6. CENTRAL
Shea TC, Kewalramani T, Mun Y, Jayne G, Dreiling LK. Evaluation of single‐dose palifermin to reduce oral mucositis in fractionated total‐body irradiation and high‐dose chemotherapy with autologous peripheral blood progenitor cell transplantation. Journal of Supportive Oncology 2007;5(4 Suppl 2):60‐2. CENTRAL

Spielberger 2001 {published data only}

Spielberger RT, Stiff P, Emmanouilides C, Yanovich S, Bensinger W, Hedrick E, et al. Efficacy of recombinant human keratinocyte growth factor (rHuKGF) in reducing mucositis in patients with hematologic malignancies undergoing autologous peripheral blood progenitor cell transplantation (auto‐PBPCT) after radiation‐based conditioning ‐ results of a phase 2 trial. Proceedings of the American Society of Clinical Oncology 2001;20(Pt 1):7a. [Abs No 25]CENTRAL

Al‐Dasooqi 2013

Al‐Dasooqi N, Sonis ST, Bowen JM, Bateman E, Blijlevens N, Gibson RJ, et al. Emerging evidence on the pathobiology of mucositis. Supportive Care in Cancer 2013;21(11):3233‐41.

Bellm 2002

Bellm LA, Cunningham G, Durnell L, Eilers J, Epstein JB, Fleming T, et al. Defining clinically meaningful outcomes in the evaluation of new treatments for oral mucositis: oral mucositis patient provider advisory board. Cancer Investigation 2002;20(5‐6):793‐800.

Boers‐Doets 2013

Boers‐Doets CB, Raber‐Durlacher JE, Treister NS, Epstein JB, Arends AB, Wiersma DR, et al. Mammalian target of rapamycin inhibitor‐associated stomatitis. Future Oncology 2013;9(12):1883‐92.

Clarke 2007

Clarke M. Standardising outcomes for clinical trials and systematic reviews. Trials 2007;8:39.

Dwan 2008

Dwan K, Altman DG, Arnaiz JA, Bloom J, Chan AW, Cronin E, et al. Systematic review of the empirical evidence of study publication bias and outcome reporting bias. PLOS One 2008;3(8):e3081.

Egger 1997

Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta‐analysis detected by a simple, graphical test. BMJ 1997;315(7109):629‐34.

Elting 2008

Elting LS, Keefe DM, Sonis ST, Garden AS, Spijkervet FK, Barasch A, et al. Patient‐reported measurements of oral mucositis in head and neck cancer patients treated with radiotherapy with or without chemotherapy: demonstration of increased frequency, severity, resistance to palliation, and impact on quality of life. Cancer 2008;113(10):2704‐13.

Epstein 1999

Epstein JB, Emerton S, Kolbinson DA, Le ND, Phillips N, Stevenson‐Moore P, et al. Quality of life and oral function following radiotherapy for head and neck cancer. Head & Neck 1999;21(1):1‐11.

GRADE 2004

Atkins D, Best D, Briss PA, Eccles M, Falck‐Ytter Y, Flottorp S, et al. Grading quality of evidence and strength of recommendations. BMJ 2004;328(7454):1490.

Higgins 2011

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

Jensen 2014

Jensen SB, Peterson DE. Oral mucosal injury caused by cancer therapies: current management and new frontiers in research. Journal of Oral Pathology and Medicine 2014;43(2):81‐90.

Lalla 2008

Lalla RV, Sonis ST, Peterson DE. Management of oral mucositis in patients who have cancer. Dental Clinics of North America 2008;52(1):61‐77.

Lalla 2014

Lalla RV, Bowen J, Barasch A, Elting L, Epstein J, Keefe DM, et al. MASCC/ISOO clinical practice guidelines for the management of mucositis secondary to cancer therapy. Cancer 2014;120(10):1453‐61.

Lefebvre 2011

Lefebvre C, Manheimer E, Glanville J. Chapter 6: Searching for studies. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Lyman 2010

Lyman GH, Dale DC, Wolff DA, Culakova E, Poniewierski MS, Kuderer NM, et al. Acute myeloid leukemia or myelodysplastic syndrome in randomized controlled clinical trials of cancer chemotherapy with granulocyte colony‐stimulating factor: a systematic review. Journal of Clinical Oncology 2010;28(17):2914‐24.

Miller 2001

Miller M, Kearney N. Oral care for patients with cancer: a review of the literature. Cancer Nursing 2001;24(4):241‐54.

Nonzee 2008

Nonzee NJ, Dandade NA, Patel U, Markossian T, Agulnik M, Argiris A, et al. Evaluating the supportive care costs of severe radiochemotherapy‐induced mucositis and pharyngitis. Cancer 2008;113(6):1446‐52.

Peterson 2015

Peterson DE, Srivastava R, Lalla RV. Oral mucosal injury in oncology patients: perspectives on maturation of a field. Oral Diseases2015; Vol. 21, issue 2:133‐41.

Raber‐Durlacher 2013

Raber‐Durlacher JE, von Bültzingslöwen I, Logan RM, Bowen J, Al‐Azri AR, Everaus H, et al. Systematic review of cytokines and growth factors for the management of oral mucositis in cancer patients. Supportive Care in Cancer 2013;21(1):343‐55.

Riley 2017

Riley P, Glenny AM, Hua F, Worthington HV. Pharmacological interventions for preventing dry mouth and salivary gland dysfunction following radiotherapy. Cochrane Database of Systematic Reviews 2017, Issue 7. [DOI: 10.1002/14651858.CD012744]

Scully 2006

Scully C, Sonis S, Diz PD. Oral mucositis. Oral Diseases 2006;12(3):229‐41.

Sonis 2001

Sonis ST, Oster G, Fuchs H, Bellm L, Bradford WZ, Edelsberg J, et al. Oral mucositis and the clinical and economic outcomes of hematopoietic stem‐cell transplantation. Journal of Clinical Oncology 2001;19(8):2201‐5.

Sonis 2004

Sonis ST, Elting LS, Keefe D, Peterson DE, Schubert M, Hauer‐Jensen M, et al. Perspectives on cancer therapy‐induced mucosal injury: pathogenesis, measurement, epidemiology, and consequences for patients. Cancer 2004;100(9 Suppl):1995‐2025.

Sonis 2009

Sonis ST. Mucositis: the impact, biology and therapeutic opportunities of oral mucositis. Oral Oncology 2009;45(12):1015‐20.

Trotti 2003

Trotti A, Bellm LA, Epstein JB, Frame D, Fuchs HJ, Gwede CK, et al. Mucositis incidence, severity and associated outcomes in patients with head and neck cancer receiving radiotherapy with or without chemotherapy: a systematic literature review. Radiotherapy and Oncology 2003;66(3):253‐62.

Turhal 2000

Turhal NS, Erdal S, Karacay S. Efficacy of treatment to relieve mucositis‐induced discomfort. Supportive Care in Cancer 2000;8(1):55‐8.

von Bültzingslöwen 2006

von Bültzingslöwen I, Brennan MT, Spijkervet FK, Logan R, Stringer A, Raber‐Durlacher JE, et al. Growth factors and cytokines in the prevention and treatment of oral and gastrointestinal mucositis. Supportive Care in Cancer 2006;14(6):519‐27.

Williamson 2005

Williamson PR, Gamble C, Altman DG, Hutton JL. Outcome selection bias in meta‐analysis. Statistical Methods in Medical Research 2005;14(5):515‐24.

Williamson 2012

Williamson PR, Altman DG, Blazeby JM, Clarke M, Devane D, Gargon E, et al. Developing core outcome sets for clinical trials: issues to consider. Trials 2012;13:132.

Worthington 2015

Worthington H, Clarkson J, Weldon J. Priority oral health research identification for clinical decision‐making. Evidence‐based Dentistry 2015;16(3):69‐71.

References to other published versions of this review

Riley 2015

Riley P, Glenny AM, Worthington HV, Littlewood A, Clarkson JE, McCabe MG. Interventions for preventing oral mucositis in patients with cancer receiving treatment: cytokines and growth factors. Cochrane Database of Systematic Reviews 2015, Issue 12. [DOI: 10.1002/14651858.CD011990]

Worthington 2011

Worthington HV, Clarkson JE, Bryan G, Furness S, Glenny AM, Littlewood A, et al. Interventions for preventing oral mucositis for patients with cancer receiving treatment. Cochrane Database of Systematic Reviews 2011, Issue 4. [DOI: 10.1002/14651858.CD000978.pub5]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Antoun 2009

Methods

Trial design: parallel (2 arms)

Location: Institut Gustave Roussy, Villejuif, France

Number of centres: 1

Study duration: February 2005 to September 2006
Trials registry number: none/unknown

Participants

Inclusion criteria: adults with metastatic colorectal adenocarcinoma (grade 3 to 4); life expectancy greater than 3 months; receiving 5FU‐based chemotherapy

Exclusion criteria: HIV; pregnant or lactating; unlikely to comply with interventions; participation in another trial in the previous 12 months (unless regarding chemotherapeutic protocols); undergone a total colectomy; state of subocclusion; chronic inflammatory diseases of the digestive tract; radiation enteropathy

Cancer type: metastatic colorectal adenocarcinoma (grade 3 to 4)

Cancer treatment: 5FU‐based chemotherapy

Age at baseline (years): median 60 (not reported by group)

Gender: not reported

Number randomised: 22 (not reported by group)

Number evaluated: 13 (Group A: 9; Group B: 4)

Interventions

Comparison: TGF‐beta(2) versus placebo

Group A: nutritional supplement of proteins, carbohydrates, fats, vitamins and minerals, with TGF‐beta(2) (2 ng/mg protein); formulas were in powder form, mixed with cool previously boiled water at 0.23 g/mL (100 kcl/100 mL); during each cycle participants received 750 mL to 1000 mL per day plus any other food desired; formula administered for 2 days before, 2 days during, and 3 days after chemotherapy (7 days/cycle)

Group B: same as above without the TGF‐beta(2)

Compliance: "Nine randomised patients who never ate the formula were excluded from the study" (not reported by group)

Duration of treatment: "3 months (test or control formula), for a minimum of one and a maximum of eight cycles of treatment"

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (no details reported on who assessed this, or when it was assessed; only reports incidence of any mucositis)

  • Chemotherapy‐induced diarrhoea (not an outcome of this review)

Notes

Sample size calculation: not reported

Funding: "This study was funded by Nestec Ltd" ‐ Nestlé (manufacturer of the intervention)

Declarations/conflicts of interest: 6 of the 9 authors were either consultants (1) or employees (5) of Nestlé

Data handling by review authors: reported in additional table

Other information of note: "Due to low accrual of patients (22 patients were enrolled and randomised in 18 months), the study was prematurely stopped"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomly assigned"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "randomly assigned"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind" and "The test formula differed only by containing an additional..."

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of oral mucositis using this scale, requiring the patient's assessment of pain/soreness and their ability to swallow but, as the participants were unaware of their group allocation, the assessment of oral mucositis can be considered to be blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall attrition was 41% although it was not reported by group

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Blazar 2006

Methods

Trial design: parallel (7 arms) dose‐ranging study

Location: Universities of Minnesota and Michigan, USA

Number of centres: 2

Study duration: not reported
Trials registry number: none/unknown

Participants

Inclusion criteria: aged 3 to 65 years; diagnosed with haematological malignancy (including myelodysplastic syndromes); ECOG score of 0 to 2; eligible for allogeneic HSCT after conditioning treatment with chemotherapy with or without TBI

Exclusion criteria: received previous allogeneic HSCT; due to receive a T‐cell‐depleted donor graft; active chronic skin disease; pre‐existent inflammatory bowel disease; uncontrolled (antibiotic‐resistant) bacterial infection; hepatitis; HIV

Cancer type: haematologic: ALL (Group A: 12%; Group B: 3%); AML (Group A: 35%; Group B: 39%); CML (Group A: 10%; Group B: 26%); MDS (Group A: 9%; Group B: 19%); NHL (Group A: 19%; Group B: 3%); Hodgkin's (Group A: 1%; Group B: 0%); Other (Group A: 14%; Group B: 10%)

Cancer treatment: both centres had allogeneic HSCT on day 0 but differed in conditioning regimen and GVHD prophylaxis as follows:

  • Minnesota centre (n = 54): conditioning with cyclophosphamide (60 mg/kg per day for 2 days) on days ‐7 and ‐6 and TBI (total dose 13.2 Gy, fractionated as 165 cGy twice daily for 4 days) on days ‐4 to ‐1; GVHD prophylaxis with methotrexate (15 mg/m², IV bolus on day +1, and 10 mg/m², IV bolus on days 3, 6, and 11) and cyclosporine A (starting from day ‐3); Group A: 69%; Group B: 31%

  • Michigan centre (n = 46): conditioning with busulfan (1 mg/kg per dose given 4 times daily for 4 days) on days ‐8 to ‐5 and cyclophosphamide (60 mg/kg per day for 2 days) on days ‐3 and ‐2; GVHD prophylaxis with methotrexate (15 mg/m², IV bolus on day +1, and 10 mg/m², IV bolus on days 3, 6, and 11) and tacrolimus or cyclosporine A (starting from day ‐3); Group A: 70%; Group B: 30%

Both centres received G‐CSF (filgrastim) 5 µg/kg per day from 24 hours after HSCT until neutrophil recovery

Age at baseline (years): Group A: median 46 (range 7 to 65); Group B: median 46 (range 7 to 63)

Gender: both groups 58% male

Number randomised: 100 (Group A: 69; Group B: 31)

Number evaluated: 96 (Group A: 65; Group B: 31)

Interventions

Comparison: KGF (palifermin) versus placebo

(4 KGF arms and 3 placebo arms were each combined into a single arm)

Group A: KGF

  • (n = 8): 40 µg/kg per day in 6 doses on days ‐11, ‐10, ‐9 and 0, 1, 2 (total dose = 240 µg/kg)

  • (n = 10): 60 µg/kg per day in 6 doses on days ‐11, ‐10, ‐9 and 0, 1, 2 (total dose = 360 µg/kg)

  • (n = 14): 60 µg/kg per day in 9 doses on days ‐11, ‐10, ‐9 and 0, 1, 2 and 7, 8, 9 (total dose = 540 µg/kg)

  • (n = 37): 60 µg/kg per day in 12 doses on days ‐11, ‐10, ‐9 and 0, 1, 2 and 7, 8, 9 and 14, 15, 16 (total dose = 720 µg/kg)

Group B: placebo with matching schedule to either the 6, 9 or 12 dose regimen

Mode of administration not described but presumably IV as in other KGF studies

Compliance: Group A: 20 did not receive all study doses (17 of these were replaced to allow a full assessment of safety); Group B: 2 did not receive all study doses (1 replaced)

Duration of treatment: varied from 13 days (6 doses) to 27 days (12 doses) ‐ see above

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (measured 3 times per week during hospitalisation by designated observers, maximum score reported)

  • Adverse effects (assessed daily during study period using WHO and NCI‐CTC toxicities scale)

  • Incidence and severity of acute GVHD (not an outcome of this review)

  • Overall survival (not an outcome of this review)

  • Incidence of transplantation‐related toxicity (not an outcome of this review)

  • Time to marrow engraftment (not an outcome of this review)

Notes

Sample size calculation: not reported

Funding: government grants from NIH and FDA, and also supported by Amgen (pharmaceutical industry)

Declarations/conflicts of interest: not reported

Data handling by review authors: the data for incidence of mucositis were not reported separately for each dose and therefore it was not possible to include head‐to‐head comparisons of different dosages in this review; the data for incidence of mucositis were presented in subgroups of those that did or did not receive the final methotrexate infusion on day 11 but we used the overall data in our meta‐analyses (the study authors report that there was no difference between these subgroups)

Other information of note: the study authors report a greater decrease in incidence of grade 3 to 4 (severe) oral mucositis due to palifermin in the Minnesota participants (who received a more mucotoxic conditioning regimen) than in the Michigan participants

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "patients...were randomly assigned..."

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "patients...were randomly assigned..."

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of oral mucositis using this scale, requiring the patient's assessment of pain/soreness and their ability to swallow but, as the participants were unaware of their group allocation, the assessment of oral mucositis can be considered to be blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "Data for all patients randomly assigned and who received a transplant were used in all other analyses (intent‐to‐treat)"

Comment: although 18 participants (Group A: 17; Group B: 1) were replaced to allow a full assessment of safety, it seems that the originally randomised participants were included in the analyses

Overall attrition was 4% (Group A: 6%; Group B: 0%) for the oral mucositis incidence outcome. The reasons were unclear but this proportion of attrition is unlikely to have biased the results

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Blijlevens 2013

Methods

Trial design: parallel (3 arms)

Location: Europe (Italy, France, the Netherlands, Ireland, Germany, UK, Denmark, Austria, Switzerland, Czech Republic, Sweden, Hungary, Belgium, Finland)

Number of centres: 39

Study duration: December 2006 to February 2009
Trials registry number: NCT00434161

Participants

Inclusion criteria: aged between 18 and 70 years; due to receive high‐dose melphalan; ECOG score of 0 to 2 (or 3, if reason was due to multiple myeloma); at least 2 x 10⁶ CD34+ cells per kg; corrected carbon monoxide diffusing capacity 50% or higher of predicted; absolute neutrophil count at least 1.5 x 10⁹/L and platelets at least 100 x 10⁹/L; total bilirubin 2 mg/dL or lower; aspartate aminotransferase and/or alanine aminotransferase 4 x institutional upper limit of normal or lower

Exclusion criteria: not reported

Cancer type: multiple myeloma

Cancer treatment: 1‐day administration of high‐dose melphalan (200 mg/m²) on day ‐2, followed by auto‐SCT on day 0

Age at baseline (years): Group A: median 55 (range 32 to 69); Group B: median 58 (range 40 to 68); Group C: median 58 (range 41 to 68)

Gender: Group A: 54% male; Group B: 55% male; Group C: 58% male

Number randomised: 281 (Group A: 109; Group B: 115; Group C: 57)

Number evaluated: 281 (Group A: 109; Group B: 115; Group C: 57)

Interventions

Comparison: KGF versus placebo

Group A: KGF (60 µg/kg) daily IV on days ‐6, ‐5, and ‐4, then placebo on days 0 (the day of auto‐SCT), 1, and 2 (total dose = 180 µg/kg)

Group B: KGF (60 µg/kg) daily IV on days ‐6, ‐5, ‐4, 0, 1, and 2 (total dose = 360 µg/kg)

Group C: placebo daily IV on days ‐6, ‐5, ‐4, 0, 1, and 2

Compliance: Group A: 8% discontinued; Group B: 12% discontinued; Group C: 4% discontinued; (point of discontinuation or number of treatments not stated for any group)

Duration of treatment: 6 treatment days (over 9 days)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed daily by nurses and physicians from day ‐2 to day 32, maximum score reported) (duration of grade 2 to 4 and 3 to 4 oral mucositis also measured but not outcomes of this review)

  • Oral pain: OMDQ 5‐point scale for mouth and throat soreness (higher = worse pain) (assessed daily by participants from day ‐2 to day 32, data reported as AUC, not used)

  • Quality of life: EQ‐5D 0 (worst imaginable health) to 10 (best imaginable health) scale, incorporating mobility, self‐care, usual activities, pain/discomfort and anxiety/depression (assessed daily by participants from day ‐2 to day 32, mean reported only at day 7 and with no SD, no usable data)

  • Normalcy of diet (measured as incidence of TPN) (duration of TPN also measured but not used for analysis in review)

  • Adverse events (NCI‐CTC version 3.0 toxicity scale)

  • Number of days in hospital

  • Number of days of treatment with opioid analgesics (incidence of opioid analgesic use also measured and reported but not an outcome of this review)

  • Febrile neutropenia (not an outcome of this review)

  • Significant infections (not an outcome of this review)

  • Anti‐infective (IV) drug use (not an outcome of this review)

  • Blood product use (not an outcome of this review)

  • Nonopioid analgesic use (not an outcome of this review)

Notes

Sample size calculation: 275 participants required at 95% power and 5% significance to detect an odds ratio of at least 3.5 between placebo and KGF in grade 2 to 4 oral mucositis

Funding: sponsored by Swedish Orphan Biovitrum (pharmaceutical industry); KGF and placebo manufactured and packaged by Amgen (pharmaceutical industry)

Declarations/conflicts of interest: 2 authors were employees of the sponsors; the remaining authors declared no competing financial interests

Data handling by review authors: we combined the 2 KGF groups to make a single pairwise comparison against placebo; we also made a separate comparison of the 2 KGF regimens

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomization was performed by using an interactive‐voice‐response‐system before planned admission"

Comment: large multicentre trial using high‐tech randomisation method ‐ likely to be done properly

Allocation concealment (selection bias)

Low risk

Quote: "Randomization was performed by using an interactive‐voice‐response‐system before planned admission"

Comment: large multicentre trial using high‐tech randomisation method ‐ likely to be done properly

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "Study drug...packaged...in identical vials"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of lower grades of oral mucositis using the WHO scale, requiring the patient's assessment of pain/soreness and their ability to swallow. Higher grades have more objective elements so may not be affected by potential lack of blinding of the assessor. This would be the same for other subjective and objective outcomes

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately (although quality of life reported with no SD or P values, this does not affect the risk of bias judgement for other outcomes)

Other bias

Low risk

No other sources of bias are apparent

Bradstock 2014

Methods

Trial design: parallel (2 arms)

Location: Australia

Number of centres: 23

Study duration: recruitment from September 2006 to April 2010
Trials registry number: ACTRN012605000095662 (mentioned in trial report)

Participants

Inclusion criteria: aged 15 to 60 years with newly diagnosed and previously untreated (except for hydroxycarbamide for high presenting white blood cell count) acute myeloid leukaemia ‐ all subtypes except t(15;17) or variants, or core‐binding factor AML (t(8;21) or inv(16) or variants); ECOG score of 0 to 3; no history of cancer (other than basal cell skin cancer or carcinoma of the cervix in situ, or other localised cancer treated by surgical excision only more than 5 years earlier without evidence of recurrence in the intervening period)

Exclusion criteria: not reported

Cancer type: acute myeloid leukaemia

Cancer treatment: induction chemotherapy consisting of: idarubicin 9 mg/m² daily IV infusion on days 1 to 3; etoposide 75 mg/m² daily IV infusion on days 1 to 7; cytarabine 3 g/m² 12‐hourly IV infusion on days 1, 3, 5, and 7

All participants received G‐CSF (pegfilgrastim) 6 mg subcutaneously on day 8

Age at baseline (years): Group A: mean 46 (SD 12; range 17 to 60); Group B: mean 44 (SD 12; range 16 to 60)

Gender: Group A: 61% male; Group B: 67% male

Number randomised: 160 (Group A: 79; Group B: 81)

Number evaluated: 151 (Group A: 73; Group B: 78)

Interventions

Comparison: KGF (palifermin) versus placebo

Group A: KGF (60 µg/kg) daily IV on days ‐3, ‐2, ‐1 prior to chemotherapy and for 3 days after completion of chemotherapy (total dose = 360 µg/kg)

Group B: same schedule with placebo

Compliance: received all 3 pre‐chemotherapy doses: Group A: 97%; Group B: 100%; received all 3 post‐chemotherapy doses: Group A: 95%; Group B: 96%

Duration of treatment: 6 treatment days (over 14 days)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed daily by investigators and specifically trained site personnel from the first day of chemotherapy and until the earlier of the date of discharge or day 28 after the start of chemotherapy, maximum score reported) (duration of grade 3 to 4 oral mucositis also measured but not an outcome of this review)

  • Adverse events (NCI‐CTC version 2.0 toxicity scale)

  • Incidence of severe gastrointestinal toxicities related to the induction chemotherapy (not an outcome of this review)

  • Complete response to chemotherapy (not an outcome of this review)

Notes

Sample size calculation: 128 per group required to detect a reduction in grade 3 to 4 mucositis from 22% to 10% at 70% power and 5% significance

Funding: "This study was funded in part from Project Grant 302133 from the National Health and Medical Research Council of Australia" (government); KGF and placebo provided by Amgen (pharmaceutical industry)

Declarations/conflicts of interest: "The authors have no conflicts of interest to declare"

Data handling by review authors: N/A

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Eligible patients were randomized 1:1 using a block randomization technique and stratification by participating centre to receive placebo or palifermin"

Comment: large multicentre trial using block randomisation and stratification ‐ likely to be done properly

Allocation concealment (selection bias)

Unclear risk

Quote: "Eligible patients were randomized 1:1 using a block randomization technique and stratification by participating centre to receive placebo or palifermin"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "placebo‐controlled" and "Both investigators and patients were blinded to the randomization outcome"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "placebo‐controlled" and "Both investigators and patients were blinded to the randomization outcome"

Comment: investigators assessed oral mucositis, which would have partly relied on patient's assessment of pain/soreness and their ability to swallow; both investigators and patients were blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall attrition was 6% (Group A: 8%; Group B: 4%) for the oral mucositis incidence outcome. The reasons were similar between groups and this proportion of attrition is unlikely to have biased the results

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Brizel 2008

Methods

Trial design: parallel (2 arms)

Location: Australia, Canada and USA

Number of centres: 22

Study duration: September 1999 to May 2001
Trials registry number: none/unknown

Participants

Inclusion criteria: adults with newly diagnosed stage III/IVa or IVb squamous carcinoma of the oral cavity, oropharynx, nasopharynx, hypopharynx, and larynx (or with unknown primary and extensive neck disease) undergoing CRT intended to be curative; Karnofsky performance score of 60 or higher; haemoglobin 10 g/dL or higher; white blood cell count 3.5 x 10⁹/L or higher or absolute neutrophil count 1.5 x 10⁹/L or higher; platelet count 100 x 10⁹/L or higher; serum bilirubin 1.5 mg/dL or lower; serum creatinine lower than 2.0 mg/dL (plus a 24‐hour urinary creatinine clearance 50 mL/min in those aged 60 years or older)

Exclusion criteria: previous RT to the head and neck; previous surgery for the primary tumour (not including biopsy); previous CT; allergy to Escherichia coli‐derived products; participation in any other investigational study within the 30 days prior to this study; refusal to use adequate contraception during the study; pregnant or breastfeeding

Cancer type: head and neck: oral (Group A: 12%; Group B: 6%); oropharynx/nasopharynx (Group A: 61%; Group B: 66%); hypopharynx/larynx (Group A: 27%; Group B: 28%)

Cancer treatment:

  • Radiotherapy: standard (once daily 2‐Gy fractions, 5 days per week; total 70 Gy over 7 weeks) or hyperfractionated (twice daily 1.25‐Gy fractions with 6‐hour interval, 5 days per week with an 8 to 9 day break after 3 weeks; total 72 Gy over 6.5 weeks)

  • Chemotherapy: cisplatin (20 mg/m²/day) as IV bolus injection and 5FU (1000 mg/m²/day) as continuous infusion on the first 4 days of the first and fifth weeks of radiotherapy

Age at baseline (years): Group A: mean 54 (SD 10; range 25 to 80); Group B: mean 56 (SD 10; range 42 to 75)

Gender: Group A: 82% male; Group B: 84% male

Number randomised: 101 (Group A: 69; Group B: 32)

Number evaluated: 97 (Group A: 65; Group B: 32)

Interventions

Comparison: KGF (palifermin) versus placebo

Group A: KGF 60 µg/kg once weekly by IV bolus injection starting on the Friday before CRT began (on the following Monday), then each Friday after completion of RT for 7 weeks, and 2 more doses after completion of CRT i.e. 10 doses in total (total dose = 600 µg/kg)

Group B: same schedule with matching placebo

Compliance: 99 participants (Group A: 67; Group B: 32) received at least 1 dose of their allocated intervention; 69 participants completed the full course (Group A: 47; Group B: 22); mean number of doses (Group A: 8.4; Group B: 9.1)

Duration of treatment: 9 weeks (10 doses)

Outcomes

  • Oral mucositis: NCI‐CTC (2.0) 0 to 4 scale (measured weekly by a radiation oncologist for the first 12 weeks, reported as incidence of grade 2 to 4 i.e. moderate to severe, and grade 3 to 4 i.e. severe) (duration, time to onset, and cumulative radiotherapy dose at onset of grade 2+ and 3+ oral mucositis also measured but not outcomes of this review)

  • Interruptions to cancer treatment (unscheduled radiotherapy breaks: reported as any breaks and breaks longer than 4 days)

  • Normalcy of diet (measured as incidence of supplemental nutrition by a gastrostomy tube)

  • Adverse effects (reports collected throughout the 20‐week study)

  • Opioid analgesic use (reported as incidence; number of days of treatment with opioid analgesics is an outcome of this review and therefore we did not use these data)

  • Dysphagia (not an outcome of this review)

  • Xerostomia (not an outcome of this review)

  • Antibiotic use (not an outcome of this review)

  • Tumour response rate (not an outcome of this review)

  • Progression‐free and overall survival (assessed in a longer‐term follow‐up study) (not an outcome of this review)

Notes

Sample size calculation: based on a previous study, 99 participants required to detect a 30% difference in the duration of grade 2 or higher oral mucositis with 80% power provided that the mean duration in the placebo arm was 56 days

Funding: "Supported by Amgen Inc" (pharmaceutical industry)

Declarations/conflicts of interest: multiple and involving: employment or leadership positions with the funders (Amgen); consultant or advisory roles with the funders and other pharmaceutical companies; stock ownership with the funders; honoraria from the funders and other pharmaceutical companies; research funding from the funders and other pharmaceutical companies

Data handling by review authors: the data for incidence of mucositis were presented in subgroups of those that received standard or hyperfractionated RT but we used the overall data in our meta‐analyses; for the interruptions to radiotherapy outcome, we used the data for breaks longer than 4 days as these could be pooled with other studies in this comparison

Other information of note: the study authors report a greater decrease in incidence due to palifermin in the hyperfractionated subgroup than in the standard subgroup (see figure 3A in the study report)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were randomly assigned..."

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "Patients were randomly assigned..."

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double blinded" and "Palifermin...or matching placebo was administered by intravenous bolus"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double blinded" and "Palifermin...or matching placebo was administered by intravenous bolus"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of oral mucositis using this scale, requiring the patient's assessment of pain/soreness and their ability to swallow but, as the participants were unaware of their group allocation, the assessment of oral mucositis can be considered to be blinded. The other outcomes are objective and therefore unlikely to be affected by any potential lack of blinding of the outcome assessor(s)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall attrition was 4% (Group A: 6%; Group B: 0%) for the oral mucositis incidence outcome. The reasons were unclear but this proportion of attrition is unlikely to have biased the results

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Cartee 1995

Methods

Trial design: parallel (5 arms) dose‐ranging study

Location: Duke University Medical Centre, Durham, North Carolina, USA

Number of centres: 1

Study duration: not reported
Trials registry number: none/unknown

Participants

Inclusion criteria: premenopausal or perimenopausal patients with histologically confirmed metastatic breast cancer who had chemotherapy for inoperable or metastatic disease; performance status of 0 or 1 (CALGB criteria)

Exclusion criteria: metastatic disease involving the central nervous system; pregnant

Cancer type: stage IV breast

Cancer treatment: AFM regimen (21‐day cycle): 5FU (500 mg/m²/day) continuous infusion on days 1 to 5; adriamycin (25 mg/m²) IV bolus on days 3 to 5; methotrexate (250 mg/m²) IV on day 15 (if oral mucositis less than grade 3)

All participants received G‐CSF (filgrastim) subcutaneously (5 µg/kg/day) on days 7 to 13 and from day 16 until resolution of neutropenia

Age at baseline (years): mean 44 (not reported by group)

Gender: 49 female; 1 male (not reported by group)

Number randomised: 50 (not reported by group)

Number evaluated: 45 (Group A: 36; Group B: 9)

Interventions

Comparison: GM‐CSF (molgramostim) versus placebo

Group A: GM‐CSF

  • (n = 9 analysed): 15 mL of mouthwash (0.01 µg/mL) gently swirled in the mouth for 2 minutes before expectorating it; 4 times daily (after mealtimes, even if a meal was not eaten, and at bedtime) after routine oral care procedures; no eating/drinking for 15 minutes after using mouthwash; beginning within 24 hours of the start of 5FU and continued for the 21‐day AFM cycle (total dose = 12.6 µg)

  • (n = 9 analysed): same schedule with 0.1 µg/mL mouthwash (total dose = 126 µg)

  • (n = 9 analysed): same schedule with 1 µg/mL mouthwash (total dose = 1260 µg)

  • (n = 9 analysed): same schedule with 10 µg/mL mouthwash (total dose = 12,600 µg)

Group B: (n = 9 analysed) same schedule with matching placebo mouthwash

Compliance: (not reported by group) mouthwash therapy was discontinued if the participant experienced oral mucositis of grade 3 or above; 30 participants took at least 80% of their prescribed doses; 11 participants discontinued mouthwash therapy within 3 days prior to day 15; 4 participants discontinued mouthwash therapy between day 15 and day 21

Duration of treatment: 21 days (first treatment cycle of AFM)

Outcomes

  • Oral mucositis: CALGB 0 to 4 scale (measured on days 1 to 5, 8 to 10, 15 and 22, reported as incidence of grade 3 to 4 i.e. severe) (duration of grade 3 to 4 oral mucositis also measured but not an outcome of this review)

  • Adverse effects (assessed during study period)

  • Blood measurements (platelet, WBC, granulocyte, lymphocyte) (not an outcome of this review)

  • Myelosuppression (not an outcome of this review)

  • GM‐CSF plasma concentrations (not an outcome of this review)

Notes

Sample size calculation: this was done but the numbers required are not reported

Funding: "...supported in part by National Cancer Institute (Bethesda, MD) grant number PO1‐47741‐A4"

Declarations/conflicts of interest: not reported

Data handling by review authors: we combined the 4 GM‐CSF groups to make a single pairwise comparison against placebo and, in order to make a head‐to‐head comparison of doses, we grouped the 2 lower doses (0.01 µg/mL and 0.1 µg/mL) together and grouped the 2 higher doses (1 µg/mL and 10 µg/mL) together to make pairwise groups for comparison

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were randomized by the Duke Cancer Center Protocol Office according to a block randomization scheme and assigned a unique identifier number which designated the GM‐CSF dose level to be received"

Comment: method of sequence generation not fully described but was done by a dedicated specialist centre so was probably done adequately

Allocation concealment (selection bias)

Low risk

Quote: "Patients were randomized by the Duke Cancer Center Protocol Office" and "The patient supply of mouthwash was labelled to correspond with the assigned identifier number and dispensed by the Pharmacy. The patient assignment information was maintained by the Pharmacy"

Comment: the entire randomisation process was performed by third party so the random sequence is unlikely to have been manipulated

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "The patient assignment information was maintained by the Pharmacy"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of oral mucositis using this scale, requiring the patient's assessment of pain/soreness and their ability to swallow but, as the participants were unaware of their group allocation, the assessment of oral mucositis can be considered to be blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Overall attrition was 10%. Reasons for attrition fully reported. If all participants would have developed severe oral mucositis or dropped out due to severe oral mucositis and were all from 1 particular group, this would have biased the results. However, attrition was not reported by group, so it is unclear

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Cesaro 2013

Methods

Trial design: parallel (2 arms)

Location: Italy

Number of centres: 4

Study duration: May 2007 to June 2011
Trials registry number: EudraCT 2007‐001430‐14 (mentioned in trial report)

Participants

Inclusion criteria: aged 0 to 17 years with leukaemia, lymphoma or solid tumour; due to receive a first autologous PBSCT

Exclusion criteria: not reported

Cancer type: leukaemia/lymphoma (Group A: 22%; Group B: 17%); solid (Group A: 78%; Group B: 83%) (neuroblastoma, Ewing sarcoma/peripheral neuroectodermal tumour, medulloblastoma, Wilms tumour, central nervous system tumour)

Cancer treatment: all participants had autologous PBSCT on day 0 but differed in conditioning regimen as follows:

  • Chemotherapy: multiple regimens involving 1 to 4 chemotherapy drugs; the most common regimen was busulfan 16 mg/kg with melphalan 140 mg/m² (Group A: 53%; Group B: 37%)

  • Radiotherapy: only 4 participants (2 in each arm) had TBI 12 Gy to 14.4 Gy prior to their chemotherapy

Age at baseline (years): Group A: median 11.1 (range 1.7 to 17.4); Group B: median 11.9 (range 1.6 to 17.2)

Gender: Group A: 66% male; Group B: 59% male

Number randomised: 61 (Group A: 32; Group B: 29)

Number evaluated: 61 (Group A: 32; Group B: 29)

Interventions

Comparison: G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim)

Group A: pegfilgrastim single dose (100 µg/kg; maximum 6 mg) injected on day 3

Group B: filgrastim (5 µg/kg per day; maximum 300 µg per day) injected by 9 or more doses starting on day 3 (total dose = at least 45 µg/kg)

Mode of administration not described but presumably subcutaneously as in other G‐CSF studies

Compliance: all participants received their allocated intervention with no discontinuations

Duration of treatment: Group A: 1 day; Group B: 9 or more days

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (reported as incidence of any mucositis and grade 2 to 4 i.e. moderate to severe) (duration of any mucositis also measured but not an outcome of this review)

  • Normalcy of diet (measured as incidence of TPN) (duration of TPN also measured but not used for analysis in review)

  • Adverse events

  • Number of days in hospital (reported as median and range, unable to use data)

  • Polymorphonuclear cell recovery (not an outcome of this review)

  • Time to platelet engraftment (not an outcome of this review)

  • Incidence of febrile neutropenia and proven infection (not an outcome of this review)

  • Duration of IV antibiotics (not an outcome of this review)

  • Survival (not an outcome of this review)

Notes

Sample size calculation: based on the noninferiority of pegfilgrastim versus filgrastim in speeding the recovery of polymorphonuclear cells

Funding: "The authors have no support or funding to report"

Declarations/conflicts of interest: "The authors have declared that no competing interests exist"

Data handling by review authors: N/A

Other information of note: G‐CSF administration only began after the chemotherapy and PBSCT were completed, by which point oral mucositis may have already begun to develop

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A computer‐generated randomisation list was drawn up at Data Office Centre of AIEOP in Bologna, Italy, by a statistician not involved in patient management"

Comment: adequate method used

Allocation concealment (selection bias)

Low risk

Quote: "The list was stored by sequentially numbered sealed envelopes that was concealed to investigators until the completion of recruitment. The local investigator ...assigned each eligible patient to randomization list by phoning to AIEOP Data Office Centre"

Comment: ideal method of concealment used

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Treatment regimens were different so blinding not possible

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It would be possible to blind the outcome assessor for oral mucositis, but it was not mentioned

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Chi 1995

Methods

Trial design: cross‐over (2 arms)

Location: Cancer Centre and Department of Otolaryngology, Veterans General Hospital, Taiwan, Republic of China

Number of centres: 1

Study duration: not reported
Trials registry number: none/unknown

Participants

Inclusion criteria: diagnosed stage IV SCC of head and neck, previously untreated or locally recurrent after previous surgery or radiotherapy or both; ECOG score of 2 or above; adequate bone marrow, liver and renal function

Exclusion criteria: concurrent medical illness; local radiotherapy to oropharynx region in the previous 3 months

Cancer type: head and neck: nasopharyngeal (Group A: 44%; Group B: 27%); tongue (Group A: 22%; Group B: 36%); hypopharynx (Group A: 11%; Group B: 18%); buccal (Group A: 11%; Group B: 9%); tonsillar (Group A: 11%; Group B: 9%)

Cancer treatment: PFL regimen (21‐day cycle): cisplatin (20 mg/m²/day), 5FU (800 mg/m²/day) and leucovorin (90 mg/m²/day) IV for days 1 to 4; cycle repeated every 3 weeks (study consisted of 2 cycles)

Age at baseline (years): Group A: median 44 (range 36 to 62); Group B: median 49 (range 40 to 66)

Gender: Group A: 89% male; Group B: 91% male

Number randomised: 20 (Group A: 9; Group B: 11) ‐ figures for first cycle

Number evaluated: 20 (Group A: 9; Group B: 11)

Interventions

Comparison: GM‐CSF versus no treatment

Group A: GM‐CSF (4 µg/kg) subcutaneously from day 5 to 14 (total dose = 40 µg)

Group B: no treatment

Compliance: not reported

Duration of treatment: 10 days (days 5 to 14 of 21‐day cycle)

Outcomes

  • Oral mucositis: RTOG 0 to 4 scale (measured on days 5 to 21 by both physician's objective gross score and participant's subjective functional score, reported as area under the curve and also in the text as incidence of severe gross mucositis i.e. grade 3 to 4) (duration of grade 2 to 4 and 3 to 4 oral mucositis also measured but not an outcome of this review)

  • Adverse effects (assessed across both cycles)

Notes

Sample size calculation: not reported

Funding: "..supported in part by Department of Health, Taiwan, Republic of China, research grant no. DOH 83‐HR‐202" and "GM‐CSF (supplied by Schering Plough Corp, Kenilworth, NJ)" (pharmaceutical industry)

Declarations/conflicts of interest: not reported

Data handling by review authors: as stated in the methods section, we would only include first‐period data from cross‐over studies due to potential for period effects (which were reported in this study). The only usable data in this study were reported in the text as incidence of severe gross mucositis for the first cycle

Other information of note: the authors report a significant period effect of GM‐CSF (P < 0.01), whereby the benefits continued into the second cycle

GM‐CSF administration only began after the 4‐day chemotherapy was completed, by which point oral mucositis may have already begun to develop

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were randomized to receive GM‐CSF or no therapy"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "Patients were randomized to receive GM‐CSF or no therapy"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comparison with no treatment so blinding not possible

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It would be possible to blind the outcome assessor, as the data we used were assessed by a physician using an objective scale. However, it was not mentioned

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants appear to be included in the analyses

Selective reporting (reporting bias)

Low risk

Although most of the data were not usable in this review, this does not seem to be due to selective reporting

Other bias

Low risk

No other sources of bias are apparent

Crawford 1999

Methods

Trial design: parallel (2 arms)

Location: USA

Number of centres: 14

Study duration: recruitment from May 1988 to November 1989
Trials registry number: none/unknown

Participants

Inclusion criteria: newly diagnosed small‐cell lung cancer meeting standard criteria for end‐organ function; ECOG score of 0 to 2

Exclusion criteria: previous radiotherapy; other serious medical illnesses precluding participation

Cancer type: small‐cell lung cancer

Cancer treatment: CAE regimen (21‐day cycle): cyclophosphamide (1000 mg/m²) and doxorubicin (50 mg/m²) on day 1; etoposide (120 mg/m²) on days 1 to 3; all by IV; repeated for up to 6 cycles

Age at baseline (years): Group A: mean 61 (SD 10; range 31 to 78); Group B: mean 62 (SD 8; range 31 to 80)

Gender: Group A: 65% male; Group B: 63% male

Number randomised: 211 (Group A: 101; Group B: 110)

Number evaluated: 195 (Group A: 93; Group B: 102) ‐ figures for first cycle

Interventions

Comparison: G‐CSF (r‐metHuG‐CSF) (filgrastim) versus placebo

Group A: G‐CSF (230 µg/m²) self‐administered subcutaneously on days 4 to 17 (total dose = 3220 µg/m²)

Group B: as above but with placebo

G‐CSF stopped if postnadir neutrophil count exceeded 10 x 10⁹/L after day 12; participants kept receiving their allocated intervention until they experienced fever with neutropenia, then they received unblinded G‐CSF (230 µg/m²) in subsequent cycles; participants in the G‐CSF group who experienced fever with neutropenia were allowed 25% reduction in chemotherapy dosages in subsequent cycles

Compliance: not reported

Duration of treatment: 14 days during a 21‐day cycle

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (measured weekly, reported as incidence of any mucositis) (duration and time to onset of oral mucositis also measured but not an outcome of this review)

  • Adverse effects (assessed over the 6 cycles)

  • Number of days in hospital (reported graphically in secondary trial report with no SD or P value, no usable data)

  • Incidence, duration and severity of fever with neutropenia (not an outcome of this review)

  • Incidence and duration of antibiotic use (not an outcome of this review)

Notes

Sample size calculation: based on a difference of 20% in the incidence of fever with neutropenia over the 6 cycles

Funding: "The study was designed, coordinated, and analyzed in conjunction with Amgen, the supplier of the G‐CSF"

Declarations/conflicts of interest: not reported but some authors were employed by Amgen (pharmaceutical industry)

Data handling by review authors: for oral mucositis, we only used the data from the first cycle due to the reasons listed above (under 'Interventions')

Other information of note: G‐CSF administration only began after the 3‐day chemotherapy was completed, by which point oral mucositis may have already begun to develop.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The patients were randomly assigned to chemotherapy followed by study drug (either placebo or G‐CSF)"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "The patients were randomly assigned to chemotherapy followed by study drug (either placebo or G‐CSF)"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Placebo was supplied in matching vials for double blinding"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Placebo was supplied in matching vials for double blinding"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of oral mucositis using this scale, requiring the patient's assessment of pain/soreness and their ability to swallow but, as the participants were unaware of their group allocation, the assessment of oral mucositis can be considered to be blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall attrition was 8% (Group A: 8%; Group B: 7%) for the oral mucositis incidence outcome. The reasons were reported and similar between groups, and this proportion of attrition is unlikely to have biased the results

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Dazzi 2003

Methods

Trial design: parallel (2 arms)

Location: Ravenna, Italy

Number of centres: 1

Study duration: recruitment from July 1997 to February 2002
Trials registry number: none/unknown

Participants

Inclusion criteria: 14 years of age or older; hospitalised for high‐dose chemotherapy with autologous PBSCT

Exclusion criteria: not reported

Cancer type: breast (Group A: 17.5%; Group B: 27.5%); Ewing's sarcoma (Group A: 41.5%; Group B: 28.5%); osteosarcoma (Group A: 13%; Group B: 14%); NHL (Group A: 15%; Group B: 11.5%); germ cell tumours (Group A: 11%; Group B: 16%); small‐cell lung (Group A: 2%; Group B: 0%); soft tissue sarcoma (Group A: 0%; Group B: 2.5%)

Cancer treatment: high‐dose chemotherapy with autologous PBSCT; chemotherapy regimens were categorised into high risk and low risk and this was used as a stratification factor for randomisation, therefore high‐ and low‐risk participants were equally distributed across groups

All participants received subcutaneous G‐CSF (300 µg/day) until haematopoietic reconstitution

Age at baseline (years): Group A: median 29 (range 15 to 57); Group B: median 29 (range 17 to 61)

Gender: Group A: 59% male; Group B: 55% male

Number randomised: 90 (Group A: 46; Group B: 44)

Number evaluated: 90 (Group A: 46; Group B: 44)

Interventions

Comparison: GM‐CSF versus placebo

Group A: GM‐CSF mouthwash 150 µg/day in 100 cm³ of sterile water taken in 4 doses per day; mouthrinsing performed for 1 minute each time; treatment started on the day after the completion of chemotherapy and continued until bone marrow recovery (absolute neutrophil count > 500/mm³) or resolution of mucositis if still persistent after bone marrow recovery (total dose = variable)

Group B: as above but with placebo (sterile water)

All participants received 0.2% oral chlorhexidine and amphotericin B

Compliance: all but 7 participants regularly completed mouthwashes: 1 participant in the placebo group had none due to persistent vomiting; 6 (4 in placebo group and 2 in GM‐CSF group) started treatment but interrupted it early due to nausea and vomiting

Duration of treatment: variable and dependent on bone marrow recovery/resolution of mucositis

Outcomes

  • Oral mucositis: NCI‐CTC 0 to 4 scale (measured daily by the physicians, reported as incidence of mucositis and incidence of grade 3 to 4 i.e. severe) (duration of grade 3 to 4 oral mucositis also measured but not an outcome of this review)

  • Oral pain: 0 to 10 VAS (self‐evaluated daily, reported as mean worst score experienced)

  • Number of days of treatment with opioid analgesics (also reported as incidence; we did not use these data)

Notes

Sample size calculation: 90 participants required to detect 25% minimal difference in the rate of severe mucositis at 90% power and 5% significance

Funding: no external funding (from correspondence with authors)

Declarations/conflicts of interest: not reported

Data handling by review authors: the data for incidence of mucositis were presented in subgroups of those at low or high risk of mucositis but we used the overall data in our meta‐analyses

Other information of note: there does not appear to be any difference in risk of any or severe mucositis between the low‐ and high‐risk subgroups

GM‐CSF administration only began after the chemotherapy was completed, by which point oral mucositis may have already begun to develop

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from correspondence with authors: "The randomization list was centralized"

Comment: centralised randomisation method ‐ likely to be done properly

Allocation concealment (selection bias)

Low risk

Quote from correspondence with authors: "The randomization list was centralized"

Comment: centralised randomisation method ‐ likely to be done properly

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, randomized, placebo‐controlled study" and "The color, odor, texture and taste of both solutions were virtually identical"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, randomized, placebo‐controlled study"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of oral mucositis using this scale, requiring the patient's assessment of pain/soreness and their ability to swallow but, as the participants were unaware of their group allocation, the assessment of oral mucositis can be considered to be blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Fink 2011

Methods

Trial design: parallel (2 arms)

Location: Department of Hematology and Oncology, University Hospital Freiburg, Germany

Number of centres: 1

Study duration: March 2006 to December 2010
Trials registry number: EudraCT 2008‐001833‐87; DRKS00000043

Participants

Inclusion criteria: adults aged 18 to 75 years with either: high‐grade non‐Hodgkin's lymphoma with high‐risk syndrome (> 2 risk factors according to age‐adapted IPI = international prognostic index) in the first complete remission; Hodgkin's lymphoma in the first recurrence; recurrence of follicular lymphoma; primary therapy of a coat‐cell lymphoma (MCL) in stage II‐IV; due to receive BEAM chemotherapy followed by autologous PBSCT; Karnofsky performance score more than 60%; life expectancy more than 3 months

Exclusion criteria: previous therapy using palifermin; severe concomitant diseases with organ failure; pregnancy, lactation, positive pregnancy test; hypersensitivity to 1 of the trial drugs; severe psychiatric illness; HIV disease or immunologic deficiency; known central nervous system involvement

Cancer type: haematologic: diffuse large‐cell lymphoma (Group A: 33%; Group B: 42%); B‐cell type acute lymphocytic leukaemia (Group A: 10%; Group B: 6%); T‐cell non‐Hodgkin's lymphoma (Group A: 13%; Group B: 11%); follicular/mantle cell lymphoma (Group A: 27%; Group B: 28%); Hodgkin's lymphoma (Group A: 17%; Group B: 14%)

Cancer treatment: prior to receiving autologous PBSCT on day 0, participants received BEAM conditioning regimen from day ‐8 to ‐2: carmustine (BCNU) 300 mg/m²; etoposide 800 mg/m²; cytosine arabinoside 1600 mg/m²; melphalan 140 mg/m²

Age at baseline (years): (ITT population) Group A: median 50 (range 22 to 71); Group B: median 55 (range 22 to 73)

Gender: (ITT population) Group A: 57% male; Group B: 61% male

Number randomised: 73 (Group A: 37; Group B: 36)

Number evaluated: ITT: 66 (Group A: 30; Group B: 36); PP: 54 (Group A: 22; Group B: 32)

Interventions

Comparison: KGF (palifermin) plus best supportive care versus best supportive care alone

Group A: KGF (60 µg/kg) by IV daily for 3 days (days ‐10, ‐9, ‐8) prior to conditioning regimen and autologous PBSCT and then for 3 days after (days 0, 1, 2) (total dose = 360 µg/kg)

Group B: best supportive care ("effective oral hygiene like teeth brushing, oral rinsing") beginning on day ‐8 (the day of hospital admission for BEAM conditioning)

Compliance: Group A: 7/37 withdrew before therapy started, 3/37 had a different conditioning regimen to that specified in the study protocol (unclear if they still received KGF), 5/37 either had no KGF or did not receive all doses; Group B: 4/36 had a different conditioning regimen to that specified in the study protocol (unclear if they still received control intervention)

Duration of treatment: 6 treatment days (over 13 days)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (measured daily during hospital stay by trained nurses, study assistant, or treating physician, maximum score reported) (duration of oral mucositis also measured but not an outcome of this review)

  • Normalcy of diet (measured as incidence of TPN)

  • Adverse events

  • Number of days in hospital (medians reported, unable to use data)

  • Number of days of treatment with opioid analgesics (medians reported, unable to use data)

  • Survival (not an outcome of this review)

  • Febrile neutropenia (not an outcome of this review)

Notes

Sample size calculation: 76 participants required to detect 30% difference in the rate of severe mucositis at 80% power and 5% significance

Funding: Amgen (pharmaceutical industry)

Declarations/conflicts of interest: not reported

Data handling by review authors: data for ITT population used

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomly assigned"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "randomly assigned"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "The randomization result was known to the patient as well as to the practitioners before the start of therapy"

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote: "The randomization result was known to the patient as well as to the practitioners before the start of therapy"

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall attrition was 10% (Group A: 19%; Group B: 0%) for the ITT population. All 7 participants died before therapy started. Although this reason is not related to the outcomes, the balance created by randomisation may have been lost

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Freytes 2004

Methods

Trial design: parallel (3 arms)

Location: USA

Number of centres: 8

Study duration: not reported
Trials registry number: none/unknown

Participants

Inclusion criteria: aged 18 years or older; due to receive autologous HSCT with a conditioning regimen with a high propensity for producing mucositis (typically 50% incidence of NCI‐CTC grade 3 or 4 when receiving standard mucositis management); Karnofsky performance score of 70%; free of acute or significant chronic dental or periodontal disease at baseline examination

Exclusion criteria: previous HSCT; visible oral ulcerations at screening; pregnant or breastfeeding; childbearing potential or not using adequate contraception; history of allergy to Escherichia coli‐derived products; posterior subcapsular cataract identified at screening; history of thyroid disease prior to receiving chemotherapy (except for hypothyroidism adequately controlled with replacement therapy); history or clinical evidence of active significant acute or chronic diseases that may affect evaluation or interpretation of the effects of the study medication on mucositis; following medications: interleukin 11, topical steroids, sucralfate, hydrogen peroxide, pilocarpine, misoprostol, oral chlorhexidine rinses, or any agent that would affect the assessment of changes in the appearance of mucositis during the study

Cancer type: lymphoma (Group A: 64%; Group B: 71%; Group C: 57%); other haematologic malignancy (Group A: 36%; Group B: 29%; Group C: 43%)

Cancer treatment: prior to receiving autologous HSCT, participants received the following conditioning regimens:

  • CBV (cyclophosphamide, etoposide, and carmustine) (Group A: 50%; Group B: 64%; Group C: 43%)

  • melphalan monotherapy (Group A: 7%; Group B: 21%; Group C: 14%)

  • melphalan combination (Group A: 21%; Group B: 14%; Group C: 28%)

  • cyclophosphamide + TBI (total body irradiation) (Group A: 7%; Group B: 0%; Group C: 14%)

  • thiotepa + TBI (Group A: 7%; Group B: 0%; Group C: 0%)

  • cyclophosphamide + busulfan (Group A: 7%; Group B: 0%; Group C: 0%)

Age at baseline (years): Group A: mean 54 (SD 10); Group B: mean 47 (SD 10); Group C: mean 51 (SD 15)

Gender: Group A: 79% male; Group B: 64% male; Group C: 79% male

Number randomised: 42 (Group A: 14; Group B: 14; Group C: 14)

Number evaluated: 42 (Group A: 14; Group B: 14; Group C: 14)

Interventions

Comparison: KGF‐2 (repifermin) versus placebo

Group A: KGF‐2 (25 µg/kg) by IV daily for 3 days prior to conditioning regimen and autologous HSCT and then for 10 days after (total dose = 325 µg/kg)

Group B: KGF‐2 (50 µg/kg) as above (total dose = 650 µg/kg)

Group C: placebo as above

Compliance: not reported

Duration of treatment: 13 treatment days (over a longer period dependent on conditioning regimen)

Outcomes

  • Oral mucositis: NCI‐CTC 0 to 4 scale (incidence of grade 2, 3 or 4, assessed prior to conditioning regimen, on day of HSCT, then 3 times per week until resolution of mucositis)

  • Oral mucositis: OMAS 0 to 45 scale (reported as mean worst score and mean 3 worst scores, NCI‐CTC data used for analysis)

  • Oral and oropharyngeal pain: 0 (no pain) to 10 (worse pain) scale (assessed on days reported above, reported as mean worst score experienced)

  • Normalcy of diet: ability to eat 1 to 4 score, where 1 = normal, 2 = only soft solids, 3 = only liquids, 4 = no solids or liquids (assessed on days reported above, reported as mean worst score)

  • Adverse events (assessed from the start of the intervention until 28 days after the final dose, reported as events with a statistically significant difference between groups or that occurred in at least 50% of participants in any group and differed between groups by at least 10%)

  • Number of days of treatment with opioid analgesics (assessed as reported for oral mucositis outcome, reported as mean number of days due to mucositis pain, and mean number of days due to all pain; we used the former although acknowledge other studies typically do not specify whether or not they are reporting usage due to mucositis pain)

  • Pain on swallowing (not an outcome of this review)

  • Laboratory parameters (not outcomes of this review)

  • Immunogenicity (not an outcome of this review)

  • Electrocardiogram abnormalities, chest x‐ray assessments and ophthalmologic examinations (not outcomes of this review)

Notes

Sample size calculation: not reported

Funding: not reported

Declarations/conflicts of interest: not reported

Data handling by review authors: we combined the 2 KGF‐2 groups to make a single pairwise comparison against placebo and we also made a comparison of the 2 different KGF‐2 dosages against each other

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from correspondence with authors: "The method to implement the random allocation was by central telephone"

Comment: centralised randomisation method ‐ likely to be done properly

Allocation concealment (selection bias)

Low risk

Quote from correspondence with authors: "The method to implement the random allocation was by central telephone"

Comment: centralised randomisation method ‐ likely to be done properly

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blinded, placebo‐controlled"

Comment: the use of a placebo and identical schedule of treatment for all 3 arms should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blinded, placebo‐controlled"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of oral mucositis using the NCI‐CTC scale, requiring the patient's assessment of pain/soreness and their ability to swallow but, as the participants were unaware of their group allocation, the assessment of oral mucositis can be considered to be blinded. This would be the same for other subjective outcomes. The objective outcomes are unlikely to be affected by any potential lack of blinding of the outcome assessor(s)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Gholizadeh 2016

Methods

Trial design: parallel (2 arms)

Location: Iran

Number of centres: not reported

Study duration: not reported
Trials registry number: IRCT2013021812510N1 (mentioned in trial report)

Participants

Inclusion criteria: previously untreated acute lymphoblastic leukaemia patients; aged between 5 and 18 years

Exclusion criteria: any other systemic disease; presence of oral mucositis or other oral lesions prior to chemotherapy; history of dermatology or respiratory hypersensitivity; acute lymphoblastic leukaemia recurrence

Cancer type: acute lymphoblastic leukaemia (ALL)

Cancer treatment: induction chemotherapy protocol consisted of standard risk B‐precursor ALL (COG)/dexamethasone, vincristine, L‐asparaginase, intrathecal (methotrexate + ara‐C + hydrocortisone). The intensification protocol was dexamethasone, vincristine, L‐asparaginase/ dexamethasone, cyclophosphamide/6‐thioguanine + cytarabine + intrathecal methotrexate

Age at baseline (years): Group A: mean 8.8 (SD 2.5); Group B: mean 8.4 (SD 2.2); overall range: 5 to 18

Gender: Group A: 49% male; Group B: 49% male

Number randomised: 90 (Group A: 45; Group B: 45)

Number evaluated: 90 (Group A: 45; Group B: 45)

Interventions

Comparison: KGF (palifermin) versus chlorhexidine

Group A: KGF (60 µg/kg) by IV bolus daily for 3 days prior to chemotherapy regimen and then for 3 days after (total dose = 360 µg/kg)

Group B: chlorhexidine (concentration not reported) mouthwash used for 1 minute once daily for 3 days prior to chemotherapy regimen and then for 3 days after

Compliance: not reported

Duration of treatment: 6 treatment days (over an unspecified longer period)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (measured after 1 and 2 weeks and reported separately for each time point)

  • Adverse events

Notes

Sample size calculation: not reported

Funding: not reported

Declarations/conflicts of interest: "There is no conflict of interest in relation to this study"

Data handling by review authors: we report the data at 2 weeks as they represent the maximum oral mucositis score experienced better than those at 1 week

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The patients were randomly assigned to the palifermin or control group by using the table of random numbers"

Comment: adequate method used

Allocation concealment (selection bias)

Unclear risk

Quote: "The patients were randomly assigned to the palifermin or control group by using the table of random numbers"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comparison with chlorhexidine so blinding not possible

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Each patient was evaluated for oral lesions one and two weeks after the chemotherapy completion by the same specialist who was blind to the type of treatment" and "This limited use of chlorhexidine was to prevent the adverse effects like tooth discoloration and temporally taste changes"

Comment: grade 1 on this scale would require the unblinded participant's assessment of soreness but other aspects of the scale are more objective and were assessed by a blinded assessor. Also, blinding may not have been broken by staining/discolouration due to limited use of chlorhexidine

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Henke 2011

Methods

Trial design: parallel (2 arms)

Location: Australia, Canada, and Europe (Austria, France, Germany, Italy, Spain, UK)

Number of centres: 38

Study duration: recruitment from January 2005 to August 2007
Trials registry number: NCT00131638; 2004‐002016‐28 (EudraCT number)

Participants

Inclusion criteria: more than 18 years old; resected for pathohistologically documented high‐risk stage 2 to 4B SCC of the oral cavity, oropharynx, hypopharynx, or larynx; ECOG score of 0 to 2; at least 2 of 9 areas of the oral or oropharyngeal mucosa due to receive at least 50 Gy RT

Exclusion criteria: tumours of the lips, paranasal sinuses, salivary glands, or unknown primary site; metastatic disease; history of chronic pancreatitis or acute pancreatitis within the last year; prior RT to the head and neck region or prior chemotherapy; previous treatment on this study or with other KGFs

Cancer type: head and neck: oropharynx (Group A: 47%; Group B: 48%); oral cavity (Group A: 32%; Group B: 27%); larynx (Group A: 11%; Group B: 15%); hypopharynx (Group A: 10%; Group B: 10%); other (Group A: 1%; Group B: 1%)

Cancer treatment: after R0 or R1 resection:

  • radiotherapy: standard fractionation of once daily 2‐Gy fractions, 5 days per week; total 60 Gy (for R0 resection) over 6 weeks, or 66 Gy (for R1 resection) over 7 weeks, both with allowable range of ± 15%

  • chemotherapy: cisplatin (100 mg/m²) by IV after appropriate hydration on days 1 and 22 (for R0 resection), or days 1, 22 and 43 (for R1 resection)

Age at baseline (years): Group A: mean 56 (SD 8); Group B: mean 57 (SD 9)

Gender: Group A: 85% male; Group B: 80% male

Number randomised: 186 (Group A: 92; Group B: 94)

Number evaluated: 186 (Group A: 92; Group B: 94)

Interventions

Comparison: KGF (palifermin) versus placebo

Group A: KGF (120 µg/kg) 3 days prior to start of, and then once per week during radiochemotherapy, i.e. 7 doses for those with R0 resection, 8 doses for those with R1 resection (total dose = 840 µg/kg or 960 µg/kg respectively)

Group B: same schedule with placebo

Mode of administration not described but presumably IV as in other KGF studies

Compliance: 78% of participants in KGF group completed all planned doses compared to 86% in placebo group

Duration of treatment: 7 or 8 treatment days (over 7 or 8 weeks), depending on R0 or R1 resection respectively

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed twice weekly by trained evaluators during radiochemotherapy and then until either mucositis had reduced to grade 2 or lower or week 15, whichever occurred first, maximum score reported) (duration and time to onset of grade 3 to 4 oral mucositis also measured but not outcomes of this review)

  • Interruptions to cancer treatment: incidence of 5 or more missed consecutive RT fractions; incidence of chemotherapy delays/discontinuations

  • Oral pain: OMWQ‐HN 0 (no soreness) to 4 (extreme soreness) scale for mouth and throat soreness

  • Normalcy of diet (measured as incidence of supplemental feeding by TPN, PEG, nasogastric tube, or IV hydration) (broken down by overall supplemental feeding and also where due to oral mucositis; we used the latter although acknowledge other studies do not specify reason for supplemental feeding)

  • Adverse events: reported as those with a difference in incidence of at least 5% between arms

  • Use of opioid analgesics (total dose reported but not an outcome of this review)

  • Xerostomia (not an outcome of this review)

  • Weight change (not an outcome of this review)

  • Laboratory assessments (not an outcome of this review)

  • Survival (not an outcome of this review)

Notes

Sample size calculation: assuming 60% of placebo group would develop grade 3 to 4 mucositis, 90 per group required to detect a reduction of at least 25% at 90% power and 5% significance

Funding: "This study was supported by Amgen" (Amgen also named as sponsor on trials registry ‐ pharmaceutical industry)

Declarations/conflicts of interest: some authors had both employment or leadership positions and stock ownership within Amgen

Data handling by review authors: N/A

Other information of note: study originally randomised participants to 3 arms (180 µg/kg once per week for 7 weeks, 180 µg/kg once per week for 4 weeks followed by placebo for the next 3 doses, or placebo throughout) but, after 1 serious adverse event of respiratory insufficiency reported in 1 of the first 10 participants, the data monitoring committee decided to restart the study using 120 µg/kg doses, excluding the 17 randomised participants from the efficacy assessments. The arm with KGF for 4 weeks followed by placebo was stopped due to slow recruitment, after enrolment of 38 participants, and the results analysed in a separate appendix

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Random assignment was made by a centralized interactive voice response system"

Comment: large multicentre trial using high‐tech randomisation method ‐ likely to be done properly

Allocation concealment (selection bias)

Low risk

Quote: "Random assignment was made by a centralized interactive voice response system"

Comment: large multicentre trial using high‐tech randomisation method ‐ likely to be done properly

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, randomized, placebo‐controlled"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, randomized, placebo‐controlled"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of lower grades of oral mucositis using the WHO scale, requiring the patient's assessment of pain/soreness and their ability to swallow. Higher grades have more objective elements so may not be affected by potential lack of blinding of the assessor. This would be the same for other subjective and objective outcomes

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Hosseinjani 2017

Methods

Trial design: parallel (2 arms)

Location: Tehran University of Medical Sciences, Tehran, Iran

Number of centres: 1

Study duration: February 2014 to March 2015
Trials registry number: IRCT2015042518842N8 (mentioned in trial report)

Participants

Inclusion criteria: aged 18 years or older with non‐Hodgkin's lymphoma, Hodgkin's disease or multiple myeloma; due to receive autologous HSCT; adequate cardiac, pulmonary, renal and hepatic function

Exclusion criteria: Karnofsky performance score less than 70%; participation in another study using an unlicensed product

Cancer type: haematologic: non‐Hodgkin's lymphoma (Group A: 25%; Group B: 25%); Hodgkin's disease (Group A: 23%; Group B: 23%); multiple myeloma (Group A: 53%; Group B: 53%)

Cancer treatment: prior to receiving autologous HSCT, participants received the following conditioning regimens:

  • Hodgkin's/non‐Hodgkin's: high‐dose combination chemotherapy (carboplatin 750 mg/m² IV daily for 2 days, etoposide 300 mg/m² IV daily for 2 days, cytarabine 300mg/m²/dose IV 2 doses in each day for 2 days, and melphalan 140 mg/m² IV for 1 day)

  • Multiple myeloma: high‐dose melphalan (100 mg/m² IV daily for 2 days)

Age at baseline (years): Group A: mean 43 (SD 14); Group B: mean 45 (SD 16)

Gender: Group A: 55% male; Group B: 48% male

Number randomised: 80 (Group A: 40; Group B: 40)

Number evaluated: 80 (Group A: 40; Group B: 40)

Interventions

Comparison: Erythropoietin (recombinant human) versus placebo

Group A: 50 IU/mL erythropoietin mouthwash in aqueous vehicle (sodium benzoate, sodium citrate, citric acid, sodium hydroxide, sugar and distilled water) supplied in glass bottle stored at 4°C, 15 mL 4 times daily, starting from the first day of conditioning chemotherapy until 14 days after HSCT or until discharge from hospital (i.e. neutrophil recovery), whichever occurred first, oral intake not permitted for 1 hour following mouthwashing

Group B: same schedule with placebo (aqueous vehicle‐only)

All participants received oral hygiene care in addition to 20 drops of nystatin every 3 hours, mouthwashes containing 10 mL chlorhexidine 0.02% plus 10 mL diluted povidone iodine every 3 hours

Compliance: "However, it was a limitation of our study that EPO mouthwash administration might be affected by patients' low compliance" (no data reported)

Duration of treatment: variable and dependent on neutrophil recovery

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed daily by single trained pharmacist starting from the first day of conditioning chemotherapy and then until either 21 days after HSCT or mucositis had resolved, whichever occurred first, maximum score reported) (duration and time to onset reported but not outcomes of this review)

  • Number of days in hospital

  • Use of opioid analgesics (incidence reported but not an outcome of this review)

  • Blood measurements (not an outcome of this review)

  • Incidence and duration of fever (not an outcome of this review)

Notes

Sample size calculation: 40 per group required assuming a 30% decrease in incidence of grade 2 to 4 mucositis at 5% significance and 80% power

Funding: "There was no applicable funding source for the clinical trial"

Declarations/conflicts of interest: "The authors have no conflict of interests to report"

Data handling by review authors: there is a discrepancy in the incidence of grade 2 to 4 mucositis between Figure 2 and Table 2 (the latter has 1 extra event per group). However, this does not change the effect estimate. We have used the data in Table 2 as it reports numbers of participants along with percentages

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were randomly allocated...in a blocked randomization schedule" and "Both patient randomization and drug preparation were performed in the pharmaceutical laboratory of Pharmacy Department"

Comment: method of random sequence generation not described but done by university hospital pharmacy and therefore probably done adequately

Allocation concealment (selection bias)

Low risk

Quote: "Both patient randomization and drug preparation were performed in the pharmaceutical laboratory of Pharmacy Department"

Comment: not explicitly described but pharmacy‐controlled randomisation should have ensured concealment of the random sequence from those recruiting participants

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, randomized, placebo‐controlled" and "The study participants, the attending physician and the outcome assessor were all blind to the treatment assignment" and "There were no differences in colour, flavour, taste or container of the study drug and the placebo"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, randomized, placebo‐controlled" and "The study participants, the attending physician and the outcome assessor were all blind to the treatment assignment"

Comment: all parties were blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Jagasia 2012

Methods

Trial design: parallel (2 arms)

Location: USA (16 sites) and Australia (4 sites)

Number of centres: 20

Study duration: December 2005 to November 2008
Trials registry number: NCT00189488 (mentions obsolete number in trial report: NCT00964899)

Participants

Inclusion criteria: aged 18 years or older with haematologic malignancy (including myelodysplastic syndromes) and due to receive allogeneic SCT (marrow or PBPC) after a conditioning regimen; Karnofsky performance score of 70% or more; related donor or HLA‐matched unrelated donor identical at 6/6 HLA‐A, ‐B and ‐DRB1 loci (molecular typing of class I and class II for unrelated donors)

Exclusion criteria: other malignancies; prior SCT; previous use of KGF; active infection or oral mucositis; congestive heart failure (NYHA class III or IV); use of a T‐cell depleted graft for GVHD prophylaxis; inadequate renal, liver or pulmonary function; pregnant or breastfeeding; refusal to use adequate contraception during study; participation in another investigational device or drug trial in previous 30 days

Cancer type: haematologic: leukaemia (Group A: 71%; Group B: 79%); myelodysplastic syndrome (Group A: 16%; Group B: 12%); non‐Hodgkin's lymphoma (Group A: 12%; Group B: 8%); multiple myeloma (Group A: 0%; Group B: 1%); Hodgkin's disease (Group A: 1%; Group B: 0%)

Cancer treatment: prior to receiving allogeneic SCT on day 0, participants received 1 of the following conditioning regimens from day ‐11 to ‐2:

  • cyclophosphamide plus TBI with or without etoposide

  • TBI plus etoposide

  • melphalan plus TBI > 11 Gy

  • busulfan plus cyclophosphamide

  • busulfan plus melphalan (fully ablative doses)

  • fludarabine plus melphalan (fully ablative doses)

Participants received methotrexate (with a calcineurin inhibitor ‐ either cyclosporine or tacrolimus) for GVHD prophylaxis on days 1, 3 and 6 (planned), and on day 11 (if toxicity allowed) at doses of 15 mg/m², 10 mg/m², 10 mg/m² and 10 mg/m² respectively

Age at baseline (years): Group A: median 42 (range 18 to 62); Group B: median 44 (range 18 to 64)

Gender: Group A: 52% male; Group B: 63% male

Number randomised: 155 (Group A: 77; Group B: 78)

Number evaluated: 155 (Group A: 77; Group B: 78)

Interventions

Comparison: KGF (palifermin) versus placebo

Group A: KGF (60 µg/kg) by IV bolus daily for 3 days prior to start of conditioning therapy, then a single 180 µg/kg dose after conditioning, but often 1 or 2 days before SCT (total dose = 360 µg/kg)

Group B: same schedule with placebo

Compliance: received at least 1 dose: Group A: 99%; Group B: 96%; received all doses: Group A: 92%; Group B: 88%

Duration of treatment: 4 treatment days (over roughly 14 days)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (reported as incidence of grade 2 to 4 i.e. moderate to severe, and grade 3 to 4 i.e. severe, assessed daily by trained evaluators from day ‐11 (first day of conditioning) and then until hospital discharge or day 28, whichever occurred first) (duration also measured but not an outcome of this review)

  • Normalcy of diet (measured as incidence of TPN)

  • Adverse events

  • Use of opioid analgesics (incidence reported but not an outcome of this review)

  • Incidence and severity of acute GVHD (not an outcome of this review)

Notes

Sample size calculation: based on GVHD (not met due to early stopping)

Funding: "This study was supported by research funding from Amgen Inc. Jonathan Latham of PharmaScribe, LLC received funding from Amgen Inc. to provide assistance with the preparation of the manuscript. Xuesong Guan of Amgen Inc. provided assistance with statistical analyses" (pharmaceutical industry)

Declarations/conflicts of interest: some authors were employees and stockholders of Amgen and some received compensation from Amgen for consultation

Data handling by review authors: N/A

Other information of note: planned sample size was 200 participants but the study was stopped due to slow recruitment

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Subjects were randomly assigned..."

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "Subjects were randomly assigned..."

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: it is not clear who was blinded but grades 2 to 4 on the WHO scale are sufficiently objective and unlikely to be affected by any lack of blinding of the assessors

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Katano 1995

Methods

Trial design: parallel (2 arms)

Location: Saga Medical School, Saga, Japan

Number of centres: 1

Study duration: not reported
Trials registry number: none/unknown

Participants

Inclusion criteria: breast cancer patients

Exclusion criteria: not reported

Cancer type: breast: primary advanced (Group A: 2; Group B: 5); inflammatory (Group A: 4; Group B: 1); recurrent (Group A: 1; Group B: 1)

Cancer treatment: preoperative IA high‐dose adriamycin (10 mg to 40 mg every 2 to 3 days to a total dose of 70 mg to 170 mg)

Age at baseline (years): Group A: mean 53 (SD 11; range 38 to 69); Group B: mean 52 (SD 10; range 45 to 69)

Gender: all female

Number randomised: 14 (Group A: 7; Group B: 7)

Number evaluated: 14 (Group A: 7; Group B: 7)

Interventions

Comparison: G‐CSF versus no treatment

Group A: G‐CSF (125 µg) by daily subcutaneous injection until leukocyte counts > 8000/mm³; timing in relation to chemotherapy not specifically reported, but the group was further divided into 2 subgroups where one (n = 4) received G‐CSF during/as an adjunct to the chemotherapy, and the other (n = 3) received G‐CSF afterwards (after the leukocyte counts were likely to drop below 2000/mm³)

Group B: no treatment

Compliance: not reported

Duration of treatment: variable and dependent on leukocyte recovery (to > 8000/mm³)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed once 1 to 7 days prior to chemotherapy and then every day afterwards by a single experienced examiner, reported as incidence of grade 2 to 4) (duration of grade 2 to 4 also measured but not an outcome of this review)

  • Blood measurements (not an outcome of this review)

  • Alopecia (not an outcome of this review)

  • Adult respiratory distress syndrome (not an outcome of this review)

  • Fever (not an outcome of this review)

Notes

Sample size calculation: not reported

Funding: "G‐CSF (Neutrogin) was provided by Chugai Pharmaceutical"

Declarations/conflicts of interest: not reported

Data handling by review authors: the data for incidence of mucositis were presented in subgroups of those receiving G‐CSF during or after chemotherapy but we used the overall data in our meta‐analyses

Other information of note: both cases of mucositis were in the subgroup who received G‐CSF after chemotherapy. Oral mucositis may have already begun to develop in this subgroup

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomized into two groups"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "randomized into two groups"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comparison with no treatment so blinding not possible

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It would be possible to blind the outcome assessor, as the data we used were assessed by an examiner looking for erythema and ulcers. However, it was not mentioned

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Kim 2017

Methods

Trial design: parallel (2 arms)

Location: Seoul National University Hospital, Seoul, South Korea

Number of centres: 1

Study duration: recruitment from March 2009 to August 2014
Trials registry number: NCT00845819 (mentioned in trial report)

Participants

Inclusion criteria: aged 18 years or older with haematologic malignancy; due to receive intensive chemotherapy followed by autologous or allogeneic HSCT; normal oral cavity (grade 0 mucositis); ECOG score of 0 to 2

Exclusion criteria: received chemotherapy, radiotherapy or surgery within previous 3 weeks; history of allergy to the intervention or similar drugs; participation in other clinical trials within the previous 4 weeks with the potential to affect study results

Cancer type: haematologic: multiple myeloma (Group A: 57%; Group B: 55%); lymphoma (Group A: 36%; Group B: 38%); other (Group A: 7%; Group B: 7%)

Cancer treatment: prior to receiving autologous (Group A: 96%; Group B: 96%) or allogeneic HSCT, participants received the following conditioning regimens:

  • high‐dose melphalan (Group A: 57%; Group B: 57%)

  • mitoxantrone‐etoposide‐cytarabine‐melphalan (Group A: 15%; Group B: 23%)

  • busulfan‐etoposide‐cytarabine‐melphalan (Group A: 19%; Group B: 16%)

  • other (Group A: 9%; Group B: 4%)

Age at baseline (years): Group A: median 53 (range 18 to 65); Group B: median 51 (range 19 to 65)

Gender: Group A: 49% male; Group B: 54% male

Number randomised: 138 (Group A: 69; Group B: 69)

Number evaluated: 136 (Group A: 67; Group B: 69)

Interventions

Comparison: EGF (recombinant human) versus placebo

Group A: EGF (50 µg/mL) daily by oral spray, applied twice daily, sprayed (6 sprays per application) over the entire oral mucosa and then swallowed, no oral intake for 30 minutes afterwards; starting on first day of conditioning therapy and continuing until absolute neutrophil count recovered more than 1000 µL for 3 days and mucositis had resolved

Group B: placebo as above

Compliance: median patient compliance rate: Group A: 93% (range 35% to 100%); Group B: 92% (range 18% to 100%)

Duration of treatment: variable and dependent on neutrophil recovery/resolution of mucositis

Outcomes

  • Oral mucositis: NCI‐CTC (version 3.0) 0 to 4 scale (assessed daily during study period by researchers, reported as incidence of grade 2 to 4 i.e. moderate to severe, and grade 3 to 4 i.e. severe) (duration and time to onset reported but not outcomes of this review)

  • Oral pain: mouth and throat soreness 0 to 10 scale (reported as AUC median/range and only for those who had grade 2 to 4 mucositis ‐ data not usable)

  • Quality of life: modified OMDQ (reported as AUC median/range and only for those who had grade 2 to 4 mucositis ‐ data not usable)

  • Normalcy of diet (use of total parenteral nutrition)

  • Adverse events (NCI CTC version 3.0)

  • Number of days in hospital (listed as an outcome but not reported anywhere in the results ‐ data not usable)

  • Number of days of treatment with opioid analgesics (reported as median/range and only for those who had grade 2 to 4 mucositis ‐ data not usable)

  • Incidence of febrile neutropenia (not an outcome of this review)

  • Blood infections (not an outcome of this review)

  • Antibiotic use (not an outcome of this review)

  • Clinical laboratory measurements (not outcomes of this review)

Notes

Sample size calculation: 62 participants per group required to detect 27% difference in incidence of grade 2 to 4 mucositis with 80% power and 5% significance

Funding: multiple government grants; Daewoong Pharmaceutical Company (Seoul, Korea) only supplied interventions but provided no further funding and had no involvement with data collection, analysis or manuscript writing

Declarations/conflicts of interest: none apparent

Data handling by review authors: N/A

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were randomly assigned...using a computer‐generated randomization protocol, by the Medical Research Collaborating Center, Seoul National University Hospital"

Comment: adequate method used

Allocation concealment (selection bias)

Low risk

Quote: "Patients were randomly assigned...using a computer‐generated randomization protocol, by the Medical Research Collaborating Center, Seoul National University Hospital"

Comment: although concealment not explicitly mentioned, use of centralised/third party randomisation ‐ likely to be done properly

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "placebo‐controlled, double‐blind" and "clinicians, patients, and investigators responsible for assessing outcomes and analyzing data were masked to treatment assignments"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "placebo‐controlled, double‐blind" and "clinicians, patients, and investigators responsible for assessing outcomes and analyzing data were masked to treatment assignments"

Comment: all parties were blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 2 of 138 randomised participants were not included in the analysis

Selective reporting (reporting bias)

Low risk

Although most of the data were not usable in this review, this does not seem to be due to selective reporting

Other bias

Low risk

No other sources of bias are apparent

Le 2011

Methods

Trial design: parallel (2 arms)

Location: Canada, USA and Europe (Hungary, Poland, Austria, Germany, Italy, Czech Republic)

Number of centres: 46

Study duration: recruitment started August 2005, 4‐month follow‐up finished September 2007
Trials registry number: NCT00101582; 2005‐000213‐35 (EudraCT number)

Participants

Inclusion criteria: newly diagnosed, unresected stage 3 to 4B SCC of the oral cavity, oropharynx, nasopharynx, hypopharynx, or larynx; no evidence of secondary malignancy; at least 2 of 9 areas of the oral or oropharyngeal mucosa due to receive more than 50 Gy RT

Exclusion criteria: not reported

Cancer type: head and neck: oropharynx (Group A: 59%; Group B: 54%); oral cavity (Group A: 5%; Group B: 10%); larynx (Group A: 17%; Group B: 10%); hypopharynx (Group A: 15%; Group B: 23%); nasopharynx (Group A: 4%; Group B: 3%)

Cancer treatment:

  • Radiotherapy: standard fractionation of once daily 2‐Gy fractions, 5 days per week; total 70 Gy over 7 weeks

  • Chemotherapy: cisplatin (100 mg/m²) by IV infusion on days 1, 22 and 43

Age at baseline (years): Group A: mean 56 (SD 9); Group B: mean 55 (SD 8)

Gender: Group A: 84% male; Group B: 85% male

Number randomised: 188 (Group A: 94; Group B: 94)

Number evaluated: 188 (Group A: 94; Group B: 94)

Interventions

Comparison: KGF (palifermin) versus placebo

Group A: KGF (180 µg/kg) by IV bolus over 30 to 60 seconds, 3 days prior to start of, and then once per week during radiochemotherapy, i.e. 8 doses (total dose = 1440 µg/kg)

Group B: same schedule with placebo

Compliance: 93% (SD 19%) of planned KGF doses were administered compared to 96% (SD 14%) in placebo group

Duration of treatment: 8 treatment days (over 8 weeks)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed twice weekly by trained evaluators during radiochemotherapy and then until either mucositis had reduced to grade 2 or lower or week 15, whichever occurred first, maximum score reported) (duration and time to onset of grade 3 to 4 oral mucositis also measured but not outcomes of this review)

  • Interruptions to cancer treatment: incidence of 5 or more missed consecutive RT fractions; incidence of chemotherapy delays/discontinuations

  • Oral pain: OMWQ‐HN 0 (no soreness) to 4 (extreme soreness) scale for mouth and throat soreness (assessed twice weekly by trained evaluators during radiochemotherapy)

  • Normalcy of diet (measured as incidence of supplemental feeding by TPN, PEG, nasogastric tube, or IV hydration) (duration of supplemental feeding also reported but not used for analysis)

  • Adverse events: NCI‐CTC (version 3.0) reported separately for those related to study drugs

  • Use of opioid analgesics (total dose reported but not an outcome of this review)

  • Xerostomia (not an outcome of this review)

  • Survival (not an outcome of this review)

  • Laboratory assessments (not an outcome of this review)

  • Antipalifermin antibodies (not an outcome of this review)

Notes

Sample size calculation: assuming 60% of placebo group would develop grade 3 to 4 mucositis, 90 per group required to detect a reduction of at least 25% at 90% power and 5% significance

Funding: "Supported by Amgen" (Swedish Orphan Biovitrum named as sponsor on trials registry, Amgen named as collaborator ‐ both pharmaceutical industry)

Declarations/conflicts of interest: some authors had both employment or leadership positions and stock ownership within Amgen; some authors had received research funding from Amgen

Data handling by review authors: N/A

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A centralized randomization system assigned patients to either palifermin or placebo in a 1:1 ratio"

Comment: large multicentre trial using centralised randomisation method ‐ likely to be done properly

Allocation concealment (selection bias)

Low risk

Quote: "A centralized randomization system assigned patients to either palifermin or placebo in a 1:1 ratio"

Comment: large multicentre trial using centralised randomisation method ‐ likely to be done properly

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "placebo‐controlled, double‐blind"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "placebo‐controlled, double‐blind"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of lower grades of oral mucositis using the WHO scale, requiring the patient's assessment of pain/soreness and their ability to swallow. Higher grades have more objective elements so may not be affected by potential lack of blinding of the assessor. This would be the same for other subjective and objective outcomes

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Linch 1993

Methods

Trial design: parallel (5 arms)

Location: UK

Number of centres: 12

Study duration: recruitment from August 1989 to July 1990
Trials registry number: none/unknown

Participants

Inclusion criteria: adults due to receive BMT after conditioning

Exclusion criteria: myeloid malignancies

Cancer type: mixed haematologic and solid (not reported by group): Hodgkin's disease (29%); non‐Hodgkin's lymphoma (33%); multiple myeloma (Group A: 20%); ALL (15%); solid (3%)

Cancer treatment: prior to receiving BMT (autologous 84%; allogeneic 16%), participants received a conditioning regimen which consisted of chemotherapy only (71%) or with TBI (29%)

Age at baseline (years): median 36 (range 17 to 64) (not reported by group)

Gender: 69% male (not reported by group)

Number randomised: 121 (Group A: 96; Group B: 25); Group A represents 4 arms with different dosages

Number evaluated: 121 (Group A: 96; Group B: 25)

Interventions

Comparison: G‐CSF versus placebo

Group A: G‐CSF (2 µg/kg, 5 µg/kg, 10 µg/kg or 15 µg/kg) by 30‐minute IV daily starting from the day after BMT transplant and continuing until neutrophil count was > 1.0 x 10⁹/L for 3 consecutive days or until day 28, whichever occurred first

Group B: as above but with placebo

Compliance: not reported

Duration of treatment: variable and dependent on neutrophil recovery

Outcomes

  • Oral mucositis: unspecified scale (frequency of measurement not mentioned, no usable data)

  • Adverse events

  • Number of days in hospital (median reported, no usable data)

  • Neutropenia‐related outcomes (not outcomes of this review)

  • Antibiotic use (not an outcome of this review)

  • Fever (not an outcome of this review)

  • Sepsis (not an outcome of this review)

  • Blood product use (not an outcome of this review)

Notes

Sample size calculation: not reported

Funding: "Financial support for this trial was provided by Chugai Rhone Poulene" (pharmaceutical industry)

Declarations/conflicts of interest: 1 author was employed by the funders

Data handling by review authors: oral mucositis reported narratively in additional table

Other information of note: G‐CSF administration only began after the conditioning and BMT transplant were completed, by which point oral mucositis may have already begun to develop (although time scale of conditioning/BMT transplant not reported)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were randomised...in blocks of five by a computer‐generated randomisation schedule"

Comment: adequate method used

Allocation concealment (selection bias)

Unclear risk

Quote: "Patients were randomised...in blocks of five by a computer‐generated randomisation schedule"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "single blind...vehicle‐controlled"

Comment: the use of a placebo (the 'vehicle') should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote: "single blind...vehicle‐controlled"

Comment: the quote implies that outcome assessment was not blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

High risk

Oral mucositis not mentioned in the methods section as 1 of the study end points. It is only mentioned in the results narratively as there being no difference between any group, but with no data or P value

Other bias

Low risk

No other sources of bias are apparent

Lucchese 2016a

Methods

Trial design: parallel (2 arms)

Location: Vita‐Salute San Raffaele University Hospital, Milan, Italy

Number of centres: 1

Study duration: conducted from April 2009 to January 2015
Trials registry number: none/unknown

Participants

Inclusion criteria: children aged 7 years or older with B‐cell lineage acute lymphoblastic leukaemia; scheduled to receive autologous HSCT after a conditioning regimen; Karnofsky performance score of 70 or more; were to have at least 1.5 x 10⁶ CD34+ cells reinfused per kilogram available for transplant; adequate cardiac, pulmonary, renal and hepatic function

Exclusion criteria: not reported

Cancer type: B‐cell lineage acute lymphoblastic leukaemia

Cancer treatment: prior to receiving autologous HSCT (on day 0), participants received a conditioning regimen which consisted of TBI delivered in 8 fractions over 3 days (‐3 to ‐1) with at least 6 hours between fractions, followed by chemotherapy on day ‐1 (type and dose of radiotherapy and chemotherapy not reported)

Age at baseline (years): Group A: median 11 (range 7 to 16); Group B: median 11 (range 7 to 16)

Gender: Group A: 52% male; Group B: 44% male

Number randomised: 60 (Group A: 30; Group B: 30)

Number evaluated: 54 (Group A: 27; Group B: 27)

Interventions

Comparison: KGF (palifermin) versus placebo

Group A: KGF (60 µg/kg) by IV, on days ‐6 (3 days prior to start of conditioning regimen), ‐5 and ‐4, and on days 0 (the day of HSCT), 1, and 2 after transplant (total dose = 360 µg/kg)

Group B: same schedule with placebo

Compliance: not reported

Duration of treatment: 6 treatment days (over 9 days)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed daily by same clinician from day ‐7 to day 28, or until severe mucositis had reduced to grade 0, 1, or 2, data in text and figure 2 do not agree with data in table 3) (duration of grade 2 to 4 and 3 to 4 oral mucositis also measured but not an outcome of this review)

  • Oral pain: OMDQ 5‐point scale for mouth and throat soreness (higher = worse pain) (assessed daily by participant, reported as AUC, not used)

  • Normalcy of diet (methods states incidence of supplemental feeding by TPN but only duration is reported, yet text states enteral and table states parenteral, not used for analysis) (patient‐reported difficulty eating and drinking also assessed daily, both using OMDQ 0 (no difficulty) to 5 (unable to do) scale, but the means are reported as whole numbers, data not used)

  • Adverse events: NCI‐CTC (version 4.0)

  • Opioid analgesic use (reported as quantity per day; number of days of treatment with opioid analgesics is an outcome of this review but only medians were reported, and therefore we did not use these data)

  • GVHD incidence and severity (not an outcome of this review)

  • Fever with neutropenia (not an outcome of this review)

  • HSV incidence (not an outcome of this review)

  • Candidiasis incidence (not an outcome of this review)

  • Superinfections incidence (not an outcome of this review)

  • Blood measurements (not an outcome of this review)

Notes

Sample size calculation: numbers required not reported but was estimated at 80% power and 5% significance

Funding: "This work was performed with Departmental funding only"

Declarations/conflicts of interest: the authors report that they have no conflict of interest (supplemental material on journal website)

Data handling by review authors: we emailed the lead author June 2017 for clarification of the oral mucositis data but, until we receive a response, we are unable to use those data

Other information of note: unclear definitions of ulcerative (should be grade 2 to 4) and severe (should be grade 3 to 4): "...ulcerative OM (WHO grades
3 and 4), incidence and duration of severe OM (WHO grades 3 and 4)..."

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A centralized randomization system assigned patients either to palifermin or conventional treatment in a 1:1 ratio......The statistician gave randomization list to the pharmacy, so the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment"

Comment: randomisation 'system' used and done by a statistician, likely to be done properly

Allocation concealment (selection bias)

Low risk

Quote: "A centralized randomization system assigned patients either to palifermin or conventional treatment in a 1:1 ratio......The statistician gave randomization list to the pharmacy, so the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment"

Comment: centralised randomisation with pharmacy assigning participants to groups

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "The statistician gave randomization list to the pharmacy, so the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment. The pharmacy provided the research team with the blinded study medication"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The statistician gave randomization list to the pharmacy, so the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment. The pharmacy provided the research team with the blinded study medication"

Comment: outcome assessors were clearly blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall attrition was same in both groups (10%) with the same reason given

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Lucchese 2016b

Methods

Trial design: parallel (2 arms)

Location: Vita‐Salute San Raffaele University Hospital, Milan, Italy

Number of centres: 1

Study duration: conducted from April 2010 to January 2014
Trials registry number: none/unknown

Participants

Inclusion criteria: children aged 9 to 15 years with B‐cell lineage acute lymphoblastic leukaemia; scheduled to receive allogeneic HSCT after a conditioning regimen

Exclusion criteria: not reported

Cancer type: B‐cell lineage acute lymphoblastic leukaemia

Cancer treatment: prior to receiving allogeneic HSCT, participants received a conditioning regimen of a total of 12 Gy TBI delivered in 8 fractions of 1.5 Gy twice daily over 4 days

Age at baseline (years): Group A: median 12 (range 8 to 15); Group B: median 12 (range 8 to 15)

Gender: Group A: 50% male; Group B: 55% male

Number randomised: 46 (Group A: 24; Group B: 22)

Number evaluated: 46 (Group A: 24; Group B: 22)

Interventions

Comparison: KGF (palifermin) versus placebo

Group A: KGF (60 µg/kg) by IV, for 3 days prior to start of conditioning regimen and for 3 days after completion (total dose = 360 µg/kg)

Group B: same schedule with placebo

Compliance: not reported

Duration of treatment: 6 treatment days (over unclear period of time ‐ not fully described)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed by the clinical research team at baseline, days 1, 2, 3 (it is not clear when this is in relation to the cancer treatment or study treatment) and at the end of the cycle ‐ "All of these results were confirmed at the last follow‐up (60 days)" ‐ it is not clear how they have been confirmed if, for example, OM has resolved by 60 days; it is not clear whether or not there were multiple cycles, incidence of each grade does not add up or make sense or match the data in table 3) (duration of grade 2 to 4 and 3 to 4 oral mucositis also measured but not an outcome of this review)

  • Normalcy of diet (duration of supplemental feeding by TPN, not used for analysis) (patient‐reported difficulty eating and drinking assessed using OMDQ 0 (no difficulty) to 5 (unable to do) scale, but the means are reported as whole numbers, data not used)

  • Adverse events

  • Opioid analgesic use (number of days of treatment with opioid analgesics (morphine) reported but unclear if mean/median and no SD or P value reported; unable to use data; also reported as quantity per day but not an outcome of this review)

Notes

Sample size calculation: not reported

Funding: "This work has been performed with departmental funding only"

Declarations/conflicts of interest: "The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript"

Data handling by review authors: we emailed the lead author June 2017 for clarification of the oral mucositis data but, until we receive a response, we are unable to use those data

Other information of note: "Patients who had severe (grade 3 or 4) OM during blinded cycles received open‐label palifermin at the same dosages as in the other group. The research team assessed patients for OM at baseline before the cycle; at days 1, 2, 3 and at the end of the transplant cycle." ‐ nowhere else in the report suggests that there were multiple cycles of treatment; unclear definitions of ulcerative mucositis i.e. described correctly in one section as grades 2 to 4, but described in another section as grades 1 to 4

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "No stratification was performed and two distinct computer‐generated a randomization lists. The statistician gave both randomization lists to the pharmacy, so the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment"

Comment: adequate method done by a statistician (although it is unclear why there were 2 lists)

Allocation concealment (selection bias)

Low risk

Quote: "No stratification was performed and two distinct computer‐generated a randomization lists. The statistician gave both randomization lists to the pharmacy, so the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment"

Comment: randomisation 'system' used and done by a statistician, likely to be done properly

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "The statistician gave both randomization lists to the pharmacy, so the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment. The pharmacy provided the research team with the blinded study medication"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The statistician gave both randomization lists to the pharmacy, so the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment. The pharmacy provided the research team with the blinded study medication"

Comment: outcome assessors were clearly blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Makkonen 2000

Methods

Trial design: parallel (2 arms)

Location: Turku University Central Hospital and Helsinki University Central Hospital, Finland

Number of centres: 2

Study duration: recruitment from November 1994 to August 1996
Trials registry number: none/unknown

Participants

Inclusion criteria: scheduled to receive total target dose of at least 56 Gy to the oropharyngeal mucosa

Exclusion criteria: prior chemotherapy or radiotherapy; concurrent use of anticholinergic drugs; autoimmune thrombocytopenic purpura; WHO performance status higher than 2

Cancer type: head and neck: mobile tongue (Group A: 30%; Group B: 20%); oral cavity, other (Group A: 40%; Group B: 25%); oropharynx (Group A: 20%; Group B: 15%); nasopharynx (Group A: 5%; Group B: 10%); supraglottic larynx (Group A: 5%; Group B: 15%); hypopharynx (Group A: 0%; Group B: 15%)

Cancer treatment:

  • conventional radiotherapy (Group A: 50%; Group B: 50%): 1.9 Gy to 2 Gy daily fractions, 5 fractions per week

  • hyperfractionated radiotherapy (Group A: 50%; Group B: 50%): 1.6 Gy fractions twice per day with minimum 6 hours between fractions, to a total dose of 38.4 Gy; planned break of 9 to 12 days during which the mucosa healed in order to allow further doses to a total of 56 Gy to 73 Gy; overall treatment time was 5 to 6 weeks

Total target dose: Group A: median 65 Gy (range 56 to 68); Group B: median 66 Gy (range 58 to 73); oral surgery prior to RT: Group A: 20%; Group B: 30%; oral surgery after radiotherapy: 23 (not reported by group)

Age at baseline (years): Group A: mean 63 (SD 10; range 47 to 87); Group B: mean 59 (SD 13; range 33 to 79)

Gender: Group A: 60% male; Group B: 55% male

Number randomised: 40 (Group A: 20; Group B: 20)

Number evaluated: unclear; results presented as percentages

Interventions

Comparison: GM‐CSF (molgramostim) plus sucralfate versus sucralfate alone

Both groups rinsed mouth with sucralfate (1 g) suspension for 1 minute before swallowing, 6 times per day, starting after 1 week of RT and continued until the end of RT (including weekends and planned/unplanned treatment breaks). Both groups advised to rinse their mouths with saline solution in between the sucralfate rinses if required

Group A: after cumulative radiation dose of 10 Gy, GM‐CSF (150 µg to 300 µg ‐ dependent on body weight) by daily subcutaneous injection until the last day of RT; not given over weekends or during planned/unplanned treatment breaks; mean total dose = 3398 µg (range 300 µg to 7200 µg)

Group B: no other treatment

Oral pain was treated with anti‐inflammatory analgesics and with local anaesthetic mouthwashes (lidocaine hydrochloride, Xylocain 0.5%)

Compliance: not reported

Duration of treatment: variable and dependent on duration of RT

Outcomes

  • Oral mucositis: 0 to 2 scale where 0 = no mucositis, 1 = erythema and edema without ulcers but food intake/use of dental prosthesis was not affected, and 2 = one or more ulcers or bleeding or food intake/use of dental prosthesis was affected (assessed weekly during RT and 1 and 6 months after completion of RT, reported at start of RT and weekly for 4 weeks after; no usable data)

  • Interruptions to cancer treatment (only 2 due to mucositis but not reported by group; data not usable)

  • Oral pain: 0 to 10 VAS and also 1 to 4 scale where 1 = no pain, 2 = mild, 3 = moderate, and 4 = severe (assessed daily at midday during RT) (data not usable ‐ 1 to 4 scale reported as "no difference" and VAS reported on a graph with no SD or P values)

  • Adverse events (due to GM‐CSF, sucralfate, or both)

  • Saliva flow rates (not an outcome of this review)

  • Salivary lactoferrin (not an outcome of this review)

  • Blood measurements (not an outcome of this review)

  • Body weight (not an outcome of this review)

  • Overall survival (not an outcome of this review)

Notes

Sample size calculation: not reported

Funding: GM‐CSF and sucralfate were both provided by pharmaceutical industry (Schering‐Plough Corporation and Orion‐Farmos Pharmaceuticals respectively)

Declarations/conflicts of interest: not reported

Data handling by review authors: no usable data

Other information of note: GM‐CSF administration began after cumulative radiation dose of 10 Gy, by which point oral mucositis may have already begun to develop

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "assignment to the treatment groups was carried out via a phone call to the randomization center located at the Finnish Cancer Registry, Helsinki"

Comment: use of a specialist randomisation centre

Allocation concealment (selection bias)

Low risk

Quote: "assignment to the treatment groups was carried out via a phone call to the randomization center located at the Finnish Cancer Registry, Helsinki"

Comment: third party/remote randomisation would have ensured concealment of the random sequence from those recruiting participants

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Blinding not possible as no placebo was used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It would be possible to blind the outcome assessor for oral mucositis as the scale was fairly objective, but it was not mentioned. It would not be possible to blind oral pain assessment as this was done by the participant who was not blinded (however, we could not use the pain data so there was no bias for this outcome)

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It is not clear how many participants were included in the analyses

Selective reporting (reporting bias)

High risk

Poor reporting of oral mucositis and oral pain

Other bias

Low risk

No other sources of bias are apparent

McAleese 2006

Methods

Trial design: parallel (2 arms)

Location: Royal Marsden Hospital, London, UK

Number of centres: 1

Study duration: recruitment from September 1997 to October 2000
Trials registry number: NCT00004256

Participants

Inclusion criteria: histologically proven T1 N0 or T2 N0 glottic carcinoma; due to receive RT with a 16‐fraction 3‐week regimen; WHO performance status 0 or 1

Exclusion criteria: renal or hepatic impairment; serious infections requiring antibiotics; taking corticosteroids or likely to require them; known allergy to GM‐CSF

Cancer type: laryngeal

Cancer treatment: accelerated radiotherapy: once‐daily fractions of 3.125 Gy, to a total dose of 50 Gy in 16 fractions over 21 days

Age at baseline (years): Group A: median 60 (range 48 to 79); Group B: median 65 (range 32 to 70)

Gender: Group A: 93% male; Group B: 86% male

Number randomised: 29 (Group A: 15; Group B: 14)

Number evaluated: 29 (Group A: 15; Group B: 14)

Interventions

Comparison: GM‐CSF (molgramostim) versus no treatment

Group A: GM‐CSF (150 µg) by daily subcutaneous injection, starting on day 14 of RT and continuing for 14 days i.e. for the last week of RT and for 1 week after completion of RT (total dose = 2100 µg)

Group B: no treatment

Compliance: 2 participants did not complete their course of GM‐CSF

Duration of treatment: 14 days

Outcomes

  • Oral mucositis: RTOG 0 to 4 scale (measured weekly by 1 of 2 independent observers using physician's objective criteria (see Appendix 9), reported graphically over time and by maximum score experienced in the text) (time to mucositis healing also measured but not an outcome of this review)

  • Normalcy of diet: use of feeding tubes

  • Adverse events

  • Analgesic use ("No differences were detected in...analgesic usage" ‐ we do not know if this is number of days or whether or not opioids)

  • Dysphagia and odynophagia (not outcomes of this review)

  • Candida infection (not an outcome of this review)

  • Laryngeal edema (not an outcome of this review)

  • Moist or dry desquamation (not an outcome of this review)

  • Weight change (not an outcome of this review)

  • Skin erythema (not an outcome of this review)

Notes

Sample size calculation: 17 per group needed at 90% power and 5% significance to detect reduction from an anticipated 60% incidence of severe mucositis to 10%

Funding: not reported

Declarations/conflicts of interest: not reported

Data handling by review authors: N/A

Other information of note: imbalance in stage distribution, with more T2 patients in the GM‐CSF group. Consequently more of this group were treated with larger fields

GM‐CSF administration only began after 14 days of radiotherapy, by which point oral mucositis may have already begun to develop

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "were randomly assigned to the active or control arms"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "were randomly assigned to the active or control arms"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Blinding not possible as no placebo was used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "At each visit one of two independent observers, blinded to group allocation, scored mucositis..."

Comment: a physician's objective version of the RTOG scale was used. Use of feeding tube is also an objective outcome

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Analgesic use reported as "No differences" with no data, however mucositis fully reported

Other bias

Low risk

No other sources of bias are apparent

Meropol 2003

Methods

Trial design: parallel (7 arms)

Location: USA

Number of centres: 10

Study duration: not reported
Trials registry number: none/unknown

Participants

Inclusion criteria: 18 years or older; metastatic colon or rectal adenocarcinoma; scheduled to receive palliative 5FU and leucovorin chemotherapy; ECOG performance status 0 to 2 (ambulatory at least 50% of waking hours); life expectancy of at least 4 months; free of lesions and no recent history (within 30 days before baseline examination) of oral ulceration, herpes simplex, oral candidiasis, severe gingivitis, or the presence of active or chronic mucositis, xerostomia, or diarrhoea; absolute neutrophil count ≥ 1.5 x 109/L; platelet count ≥ 100 x 109/L; serum creatinine ≤ 2.0 mg/dL; serum bilirubin ≤ 2.0 mg/dL; serum aspartate amino transferase less than 5 times the upper limit of normal; absence of other serious concurrent medical illness

Exclusion criteria: received chemotherapy, radiotherapy, or other investigational drugs within 4 weeks of enrolment (6 weeks for chemotherapy with mitomycin or nitrosoureas); any unresolved adverse event from previous therapy; major surgery within 2 weeks before study entry; history of insulin‐dependent diabetes mellitus; pregnant or breastfeeding; of child‐bearing potential and not using adequate contraceptive precautions; previous hypersensitivity reaction to leucovorin calcium or Escherichia coli‐derived material

Cancer type: metastatic colorectal

Cancer treatment: (palliative) leucovorin 20 mg/m² by rapid IV injection, followed immediately by 5FU 425 mg/m² by rapid IV injection for 5 consecutive days on days 4 to 8 of each 28‐day cycle

Age at baseline (years): Group A: mean 62 (SD 10; range 44 to 84); Group B: mean 66 (SD 13; range 41 to 86)

Gender: Group A: 57% male; Group B: 59% male

Number randomised: 81 (Group A: 54; Group B: 27)

Number evaluated: 81 (Group A: 54; Group B: 27)

Interventions

Comparison: KGF (recombinant human) versus placebo

Group A: KGF

  • (n = 7): 1 µg/kg per day by IV bolus on days 1 to 3 (total dose = 3 µg/kg)

  • (n = 8): 10 µg/kg as above (total dose = 30 µg/kg)

  • (n = 10): 20 µg/kg as above (total dose = 60 µg/kg)

  • (n = 11): 40 µg/kg as above (total dose = 120 µg/kg)

  • (n = 8): 60 µg/kg as above (total dose = 180 µg/kg)

  • (n = 10): 80 µg/kg as above (total dose = 240 µg/kg)

Group B: placebo as above

Compliance: 3 participants stopped KGF treatment due to adverse reactions involving the skin (1 in 60 and 2 in 80 µg/kg group)

Duration of treatment: 3 days

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed on days 1, 4, 8, 15 and 28, reported as incidence of grade 2 to 4; first cycle data only) (duration of mucositis also measured but not an outcome of this review)

  • Oral pain: self‐assessed daily questionnaire including mouth and throat pain assessed on both a 5‐point ordinal scale (reported graphically, no usable data) and a 0 to 10 VAS (reported as AUC, not used)

  • Adverse events: WHO 0 to 4 scale (reported as events of any grade that differed by at least 10% between any KGF group and placebo; events were not reported if they occurred in less than 10% of KGF group; also reported as incidence of grade 3 to 4 events)

  • Blood measurements (not an outcome of this review)

Notes

Sample size calculation: not reported

Funding: "Supported by a grant from Amgen, Inc" (pharmaceutical industry)

Declarations/conflicts of interest: not reported

Data handling by review authors: we combined the 6 KGF groups to make a single pairwise comparison against placebo; in order to make a head‐to‐head comparison of doses we grouped the 3 lower doses (1 µg/kg, 10 µg/kg and 20 µg/kg) together and grouped the 3 higher doses (40 µg/kg, 60 µg/kg and 80 µg/kg) together to make pairwise groups for comparison

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "This study was a multicenter, randomized, double‐blinded, placebo‐controlled, phase I study"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "This study was a multicenter, randomized, double‐blinded, placebo‐controlled, phase I study"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blinded, placebo‐controlled"

Comment: the use of a placebo should have ensured that blinding was successful; incidence of adverse events does not appear to differ enough between groups to cause unblinding

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blinded, placebo‐controlled"

Comment: it is not clear who was blinded but the outcome is grade 2 to 4 WHO‐scale mucositis and it is unlikely that this would be affected by any lack of blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Nemunaitis 1995

Methods

Trial design: parallel (2 arms)

Location: USA and Canada

Number of centres: 7

Study duration: 1 November 1990 to 1 July 1993
Trials registry number: none/unknown

Participants

Inclusion criteria: any age; undergoing allogeneic BMT (from HLA‐identical sibling) for haematological malignancy

Exclusion criteria: due to receive methotrexate as part of GVHD prophylaxis; chronic lymphocytic leukaemia; chronic myelogenous leukaemia in blast crisis; acute lymphoblastic leukaemia or acute myelogenous leukaemia with failure at first induction or progressed beyond a second relapse; previously received cytokines; HIV; life expectancy less than 7 days

Cancer type: haematologic malignancy: lymphoid malignancy (Group A: 21%; Group B: 16%); acute myeloid malignancy (Group A: 34%; Group B: 27%); chronic myeloid malignancy (Group A: 32%; Group B: 30%); multiple myeloma (Group A: 6%; Group B: 5%); myelodysplastic syndrome (Group A: 6%; Group B: 11%); aplastic anaemia (Group A: 2%; Group B: 11%)

Cancer treatment: prior to receiving allogeneic BMT (from HLA‐identical sibling), participants received the following conditioning regimens: busulfan and cyclophosphamide with TBI (Group A: 9%; Group B: 16%) or without TBI (Group A: 32%; Group B: 34%); busulfan, cyclophosphamide, cytosine arabinoside, and methotrexate with TBI (Group A: 23%; Group B: 13%) or without TBI (Group A: 8%; Group B: 11%); cyclophosphamide and steroid with TBI (Group A: 0%; Group B: 2%); cyclophosphamide, cytosine arabinoside, and steroid with TBI (Group A: 23%; Group B: 18%); etoposide with TBI (Group A: 0%; Group B: 2%); etoposide, cytosine arabinoside, and cyclophosphamide with TBI (Group A: 2%; Group B: 0%); etoposide, cytosine arabinoside, cyclophosphamide, and asparaginase with TBI (Group A: 0%; Group B: 2%); etoposide, methotrexate, cytosine arabinoside, and steroid with TBI (Group A: 4%; Group B: 4%)

All participants received cyclosporine and methylprednisolone for GVHD prophylaxis

Age at baseline (years): Group A: mean 32; Group B: mean 34

Gender: Group A: 60% male; Group B: 54% male

Number randomised: 109 (Group A: 53; Group B: 56)

Number evaluated: 109 (Group A: 53; Group B: 56)

Interventions

Comparison: GM‐CSF (yeast‐derived recombinant human) versus placebo

Group A: GM‐CSF (250 µg/m²) by 4‐hour IV infusion starting on day 0 (the day of BMT) to day 20 (total dose = 5250 µg/m²)

Group B: as above with placebo

Compliance: Group A: 13 participants (25%) stopped their intervention early: 11 due to adverse events (1 rash, 7 bone pain, 2 acute dyspnoea, 1 seizure); 2 (with no apparent toxicity) due to participant or physician request; Group B: 9 participants (16%) stopped their intervention early: 6 due to adverse events (2 rash, 2 bone pain, 1 elevated liver enzymes, 1 infections); 2 (on day 19) due to miscalculation of the number of days to receive the intervention; 1 (on day 9) due to severe mucositis and veno‐occlusive disease of the liver

Duration of treatment: 21 days

Outcomes

  • Oral mucositis: assessment methods are not clear but reported on 0 to 4 scale which is probably WHO (assessment frequency and duration also unclear, reported in text as incidence of grade 2 to 4 and grade 3 to 4)

  • Adverse events: 0 to 4 scale which is probably WHO (reported as incidence of any grade of event and incidence of grade 3 to 4 events with a greater than 10% frequency)

  • Number of days in hospital (reported as median values; unable to use data)

  • GVHD incidence and severity (not an outcome of this review)

  • Veno‐occlusive disease of the liver (not an outcome of this review)

  • Blood measurements (not an outcome of this review)

  • Infection: bacterial, fungal and viral (not an outcome of this review)

  • Duration of IV antibiotics (not an outcome of this review)

  • Fever (not an outcome of this review)

  • Survival: overall, disease‐free, relapse (not an outcome of this review)

Notes

Sample size calculation: not reported

Funding: "..product provided by Immunex" (pharmaceutical). Also mention of "sponsoring company" which is likely to be Immunex

Declarations/conflicts of interest: not reported but 4 authors employed by Immunex

Data handling by review authors: N/A

Other information of note: GM‐CSF administration only began after the conditioning regimen, by which point oral mucositis may have already begun to develop

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Assignment to treatment was made via a randomization schema prepared by Almedica Corporation"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Low risk

Quote: "Blinded numbered vials containing placebo or rhGM‐CSF were provided to each participating centre. The pharmacists, principal investigators, patients, support care personnel and sponsoring company were blinded to the study medication for the entire course of the study"

Comment: probably done

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind placebo‐controlled" and "The pharmacists, principal investigators, patients, support care personnel and sponsoring company were blinded to the study medication for the entire course of the study"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind placebo‐controlled" and "The pharmacists, principal investigators, patients, support care personnel and sponsoring company were blinded to the study medication for the entire course of the study"

Comment: everyone involved in the study was blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

5 participants in the placebo group received cytokines off study during the first 42 days after BMT but unlikely to bias the results in a meaningful way

Peterson 2009

Methods

Trial design: parallel (3 arms)

Location: Russia

Number of centres: 9

Study duration: recruitment from August 2006 to May 2007
Trials registry number: none/unknown

Participants

Inclusion criteria: 18 years or older with colorectal cancer (stage I to IV); undergoing chemotherapy as primary cancer therapy; experienced WHO grade 2 or higher oral mucositis during first cycle of chemotherapy, but recovered prior to enrolment (i.e. grade 0); ECOG score of 0 to 2

Exclusion criteria: pregnant (or risk of pregnancy) or lactating; scheduled to receive RT to the head and neck; received other investigational drugs within 14 days of the start of the study; evidence of alcohol and drug abuse; pre‐existing conditions such as active fungal or herpetic infection

Cancer type: colorectal (stage I to IV)

Cancer treatment: all participants received 5FU (97% with leucovorin) but 1 received capecitabine; 6 participants (3 in low‐dose ITF, 3 in placebo) also received oxaliplatin; doses or regimens comparable between groups

Age at baseline (years): Group A: median 62 ; Group B: median 56 ; Group C: median 59

Gender: Group A: 42% male; Group B: 33% male; Group C: 48% male

Number randomised: 99 (Group A: 33; Group B: 33; Group C: 33)

Number evaluated: 99 (Group A: 33; Group B: 33; Group C: 33)

Interventions

Comparison: ITF (recombinant human) versus placebo

Group A: ITF (10 mg/mL) oral spray, 300 µL (3 sprays) to the oral mucosa 8 times daily, starting on the first day (day 1) of the second chemotherapy cycle for total 14 days (total dose = 336 mg)

Group B: ITF (80 mg/mL) oral spray as above (total dose = 2688 mg)
Group C: placebo oral spray as above

Compliance: patient‐reported compliance was 97%

Duration of treatment: 14 days

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed on days 0, 1, 3, 5, 7, 10, 12, 14, and 21 ± 2, maximum score reported) (duration of mucositis also measured but not an outcome of this review)

  • Oral mucositis: OMAS 0 to 45 scale (assessed on days reported above; WHO data used for analysis)

  • Adverse events (assessed on days reported above)

  • Patient daily self‐assessment: including discolouration, mouth and throat pain, preference for solid/semi‐solid food, analgesic use, and dysphagia (no usable data for the outcomes relevant to this review)

Notes

Sample size calculation: based on previous study, 80% power to detect 40% difference in incidence of WHO grade 2 or above

Funding: The GI Company (pharmaceutical)

Declarations/conflicts of interest: some authors had roles with the funding pharmaceutical company (and other companies) both compensated and uncompensated

Data handling by review authors: we combined the 2 ITF groups to make a single pairwise comparison against placebo; we also made a separate comparison of the 2 ITF dosages

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The patients were randomly assigned"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "The patients were randomly assigned"

Comment: insufficient information to determine whether or not the random sequence was adequately concealed

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of oral mucositis using this scale, requiring the patient's assessment of pain/soreness and their ability to swallow but, as the participants were unaware of their group allocation, the assessment of oral mucositis can be considered to be blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Rosen 2006

Methods

Trial design: parallel (2 arms)

Location: USA and Australia

Number of centres: 15

Study duration: not reported
Trials registry number: none/unknown

Participants

Inclusion criteria: 18 years or older with advanced (Duke's D) colon or rectal adenocarcinoma; scheduled to receive bolus 5FU and low‐dose leucovorin (Mayo regimen) as first‐line or subsequent therapy; normal oral cavity examination (no ulceration, herpes simplex, candidiasis, or severe gingivitis); ECOG score of 0 to 2; life expectancy of 4 months or more; absolute neutrophil count of 1.5 x 10⁹/L or higher; platelet count of 100 x 10⁹/L or higher; normal renal and hepatic function

Exclusion criteria: previous radiotherapy or chemotherapy within 4 weeks or major surgery within 2 weeks of study day 1; insulin‐dependant diabetes; known allergy to leucovorin; known hypersensitivity to Escherichia coli‐derived material

Cancer type: advanced colorectal (Duke's D)

Cancer treatment: low‐dose leucovorin (20 mg/m²) by IV immediately followed by 5FU (425 mg/m²) by rapid IV once daily for 5 consecutive days on days 4 to 8 of each 28‐day cycle; 2 cycles (5FU dose could be decreased by 20% during cycle 2 if moderately severe toxicities occurred in cycle 1)

Age at baseline (years): Group A: mean 65 (SD 11.2); Group B: mean 65 (SD 11.1)

Gender: Group A: 57% male; Group B: 72% male

Number randomised: 65 (not reported by group)

Number evaluated: 64 (Group A: 28; Group B: 36)

Interventions

Comparison: KGF (palifermin) versus placebo

Group A: KGF (40 µg/kg) by IV for 3 consecutive days (days 1 to 3 of each 28‐day cycle) before the start of chemotherapy (total dose = 120 µg/kg)

Group B: as above with placebo

Compliance: not reported (only reports "palifermin was generally well tolerated")

Duration of treatment: 3 days

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed on days 0, 1, 4, 8, 12, 15, and 28, maximum score reported)

  • Oral pain: patient daily self‐assessment using OMDQ 5‐point scale for mouth and throat soreness (higher = worse pain, reported as AUC, not used)

  • Adverse events

  • Diarrhoea (related to the cancer therapy ‐ not an outcome of this review)

  • Laboratory assessments (not an outcome of this review)

  • Neutropenia (not an outcome of this review)

  • Antibody assessments (not an outcome of this review)

  • Survival (not an outcome of this review)

Notes

Sample size calculation: not reported

Funding: "Supported by Amgen Inc" (pharmaceutical industry)

Declarations/conflicts of interest: all authors were linked to the funding pharmaceutical company in terms of employment, consultancy, stock ownership, honoraria, and receipt of research funding

Data handling by review authors: data were reported separately for the chemotherapy cycles 1 and 2 ‐ we used the data for cycle 1

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were randomly assigned (by center and prior chemotherapy)"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "Patients were randomly assigned (by center and prior chemotherapy)"

Comment: insufficient information to determine method of random sequence generation

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of oral mucositis using this scale, requiring the patient's assessment of pain/soreness and their ability to swallow but, as the participants were unaware of their group allocation, the assessment of oral mucositis can be considered to be blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 randomised participant was not included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Saarilahti 2002

Methods

Trial design: parallel (2 arms)

Location: Helsinki University Central Hospital, Finland

Number of centres: 1

Study duration: recruitment from October 1999 to April 2001
Trials registry number: none/unknown

Participants

Inclusion criteria: undergone radical surgery for head and neck cancer and scheduled to receive postoperative RT to a total dose of 50 Gy or more to the oral and oropharyngeal mucosa

Exclusion criteria: prior chemotherapy or RT; chronic autoimmune or inflammatory disease; WHO performance score > 2

Cancer type: head and neck: mobile tongue (Group A: 33%; Group B: 37%); floor of mouth (Group A: 19%; Group B: 16%); tonsil (Group A: 29%; Group B: 32%); oral cavity other (Group A: 19%; Group B: 16%)

Cancer treatment: mean time from radical surgery to start of RT: Group A: 39 days (range 20 to 73); Group B: 38 days (range 20 to 56); conventionally fractionated RT to a total dose of 50 Gy to 60 Gy in 2‐Gy daily fractions, 5 times weekly (on weekdays) for 5 to 6 weeks, to the primary site and locoregional lymph nodes

Age at baseline (years): Group A: median 56 (range 43 to 87); Group B: median 60 (range 24 to 72)

Gender: Group A: 57% male; Group B: 79% male

Number randomised: 40 (Group A: 21; Group B: 19)

Number evaluated: 40 (Group A: 21; Group B: 19)

Interventions

Comparison: GM‐CSF versus sucralfate

Group A: starting after a cumulative RT dose of 10 Gy (after 1 week of RT); mouth was cleaned with water, then GM‐CSF (37.5 µg per 25 mL rinse) mouthwash rinsed around the mouth for 3 minutes then swallowed, 4 times daily after meals, on RT‐days (weekdays), until the end of RT (total dose dependent on duration of RT)

Group B: as above but with sucralfate (1 g per 25 mL rinse) (total dose dependent on duration of RT)

Compliance: not reported (only reports "both mouthwashes were well tolerated, and none of the patients reported any adverse effects related to the mouthwashes")

Duration of treatment: 4 to 5 weeks (dependent on duration of RT)

Outcomes

  • Oral mucositis: RTOG 0 to 4 scale (assessed before RT, weekly during RT, and at 1, 2, and 4 weeks after RT, reported graphically as mean score over time but authors provided maximum incidence data on request)

  • Interruptions to cancer treatment (RT interruptions due to OM)

  • Oral pain: 0 (no pain) to 10 (worse pain) VAS (assessed as reported above, reported graphically as mean score over time – no mean maximum score and no SDs, unable to use data)

  • Normalcy of diet: use of PEG feeding tube

  • Adverse events

  • Incidence of hospitalisation (number of days in hospital is an outcome of this review and therefore we did not use these data)

  • Opioid analgesic use (reported as incidence; number of days of treatment with opioid analgesics is an outcome of this review and therefore we did not use these data)

  • Laboratory parameters (not outcomes of this review)

  • Use of antimycotic agents (not an outcome of this review)

  • Use of antibiotics (not an outcome of this review)

  • Weight loss (not outcomes of this review)

Notes

Sample size calculation: not reported

Funding: not reported

Declarations/conflicts of interest: not reported

Data handling by review authors: we used oral mucositis incidence (maximum score experienced per patient) data provided by the authors; all participants experienced mucositis (grade 1 or above) as they had already received a week of RT before the intervention started, therefore we felt an analysis of incidence of any grade of mucositis should not be included

Other information of note: GM‐CSF/sucralfate administration only began after 1 week of RT, by which point oral mucositis may have already begun to develop

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomization was done using computer‐generated random digits"

Comment: adequate method used

Allocation concealment (selection bias)

Low risk

Quote: "they were assigned to a treatment group by way of a telephone call to the randomization office"

Comment: third party/remote randomisation would have ensured concealment of the random sequence from those recruiting participants

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind" and "Both solutions looked alike, and neither the investigators nor the patients were aware of the contents of the solutions given. The drug vials were marked with a study code that prevented identification of the allocation group"

Comment: adequate methods to ensure blinding

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blinded, placebo‐controlled"

Comment: nobody involved in the study was aware of group allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Schneider 1999

Methods

Trial design: parallel (2 arms)

Location: USA

Number of centres: not reported

Study duration: recruitment from January 1995 to April 1996
Trials registry number: none/unknown

Participants

Inclusion criteria: 18 years or older with histologically proven head and neck malignancy; mucosa of the oropharynx and/or oral cavity to be included in RT portal; Karnofsky performance score of 60 or more; no known hypersensitivity to E coli‐derived proteins; haemoglobin and platelet counts 100 x 10⁹/L or higher; absolute neutrophil count higher than 1.5 x 10⁹/L; normal blood pressure; agreement to bring dentition to acceptable level prior to RT (teeth with periapical abscesses or severe periodontal disease extracted, followed by a 10‐day healing period allowance)

Exclusion criteria: lactating/pregnant/females not taking effective form of contraception; scheduled to receive chemotherapy or radiosensitising drugs during the planned RT; history of myeloid malignancy; underlying collagen vascular disease; active rheumatoid arthritis

Cancer type: head and neck: nasopharynx (Group A: 0; Group B: 1); oropharynx (Group A: 2; Group B: 1); tongue (Group A: 2; Group B: 1); larynx (Group A: 2; Group B: 1); other/unknown (Group A: 2; Group B: 2)

Cancer treatment: radiotherapy in 1.8 Gy to 2 Gy standard daily fractions from Monday to Friday, to total dose 50 Gy or more

Age at baseline (years): not reported

Gender: Group A: 100% male; Group B: 83% male

Number randomised: 14 (Group A: 8; Group B: 6)

Number evaluated: 14 (Group A: 8; Group B: 6)

Interventions

Comparison: G‐CSF (r‐metHuG‐CSF) (filgrastim) versus placebo

Group A: G‐CSF (starting at 3 µg/kg and titrated to keep neutrophil count between 10 x 10⁹/L and 30 x 10⁹/L) by daily subcutaneous injection, starting on the first day of RT (weekdays), until the end of RT (total dose dependent on duration of RT and neutrophil counts)

Group B: placebo as above

Compliance: not reported (only reports "Filgrastim was well tolerated")

Duration of treatment: variable and dependent on duration of RT

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed on weekly by single examiner, reported as incidence of any mucositis and grade 3 or higher)

  • Oral mucositis: Hickey 0 to 3 scale (not used)

  • Interruptions to cancer treatment (delays or dose reductions, no usable data)

  • Weight change (not an outcome of this review)

  • Blood measurements (not an outcome of this review)

Notes

Sample size calculation: 54 required to detect 30% decrease in incidence of grade 2 or 3 mucositis at 80% power at the 5% significance level

Funding: "Financial support was provided through a grant from Amgen Inc" (pharmaceutical industry)

Declarations/conflicts of interest: not reported

Data handling by review authors: interruptions to cancer treatment outcome only reported narratively

Other information of note: study was stopped after an interim analysis. Authors state that "owing to administrative obstacles, completion of the trial is not possible", rather than due to previously stated early stopping rules

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The patients were randomized equally between the two treatment groups" and "The study material and randomization list were held by the UCLA Pharmacy for the duration of the study"

Comment: method of random sequence generation not described but done by UCLA Pharmacy and therefore probably done adequately

Allocation concealment (selection bias)

Low risk

Quote: "Amgen Inc...prepared and packaged all drug and placebo in identical containers, with the only designator being the randomization number. The study material and randomization list were held by the UCLA Pharmacy for the duration of the study"

Comment: pharmacy‐controlled randomisation would have ensured concealment of the random sequence from those recruiting participants

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "Amgen Inc...prepared and packaged all drug and placebo in identical containers, with the only designator being the randomization number"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "A single examiner...who was blinded to the patients' randomization status...scored mucositis on a weekly basis"

Comment: both participants and outcome assessor were blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

Although the study reports an interim analysis only, the study was stopped due to administrative reasons and is unlikely to introduce bias

Spielberger 2004

Methods

Trial design: parallel (2 arms)

Location: USA

Number of centres: 13

Study duration: recruitment from March 2001 to October 2002
Trials registry number: NCT00041665

Participants

Inclusion criteria: 18 years or older; Karnofsky performance score of 70 or more; scheduled to undergo autologous HSCT after conditioning regimen of fractionated TBI plus etoposide and cyclophosphamide for haematological cancers; adequate cardiac, pulmonary, renal, and hepatic function

Exclusion criteria: not reported

Cancer type: haematologic: NHL (Group A: 68%; Group B: 65%); Hodgkins (Group A: 20%; Group B: 22%); multiple myeloma (Group A: 10%; Group B: 8%); leukaemia (Group A: 2%; Group B: 5%)

Cancer treatment: prior to autologous HSCT, participants received the following conditioning regimen:

  • Radiotherapy: prior to chemotherapy, TBI of total 12 Gy in 6, 8, or 10 fractions over 3 or 4 days, with at least 6 hours between fractions

  • Chemotherapy: IV etoposide (60 mg/kg) the day after the last RT fraction (4 days before HSCT) and cyclophosphamide (100 mg/kg) 2 days later (2 days before HSCT)

All participants received G‐CSF (filgrastim) 5 µg/kg per day from HSCT until neutrophil recovery

Age at baseline (years): Group A: median 48 (range 18 to 69); Group B: median 49 (range 19 to 68)

Gender: Group A: 56% male; Group B: 68% male

Number randomised: 214 (Group A: 107; Group B: 107)

Number evaluated: 212 (Group A: 106; Group B: 106)

Interventions

Comparison: KGF (palifermin) versus placebo

Group A: KGF (60 µg/kg) by IV for 3 consecutive days starting 3 days before RT, followed by 3 more doses after HSCT starting on the day of HSCT (total dose = 360 µg/kg)

Group B: placebo as above

Compliance: 212 participants received at least 1 dose of their allocated intervention and 205 participants (Group A: 103; Group B: 102) "completed the study" (it is not clear whether or not this means that they received all doses)

Duration of treatment: 6 treatment days over 13 to 14 days

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed daily by trained assessors starting 8 days before HSCT and for 28 days after HSCT or until severe mucositis had returned to grade 0 to 2, maximum score reported) (duration of grade 3 to 4 oral mucositis also measured but not an outcome of this review)

  • Oral mucositis: RTOG 0 to 4 scale (assessed as above but only used for reporting duration of severe mucositis)

  • Oral mucositis: WCCNR 0 to 3 scale (assessed as above but only used for reporting duration of severe mucositis)

  • Oral pain: patient daily self‐assessment (day ‐12 to 28) using OMDQ 5‐point scale for mouth and throat soreness (higher = worse pain, reported as AUC, not used)

  • Quality of life: physical, functional, emotional, and social/family well‐being domains of the FACT general questionnaire (days ‐12, ‐1, 7, 10, 14, and 28, reported as AUC, not used)

  • Normalcy of diet (measured as incidence of TPN)

  • Adverse events: 1 (mild) to 5 (fatal) scale

  • Number of days of treatment with opioid analgesics (reported as median and range, unable to use data)

  • Dysphagia (not an outcome of this review)

  • Febrile neutropenia (not an outcome of this review)

  • Infections (not an outcome of this review)

  • Survival (not an outcome of this review)

  • Laboratory results (not an outcome of this review)

Notes

Sample size calculation: 210 participants required to detect a mean difference in duration of severe oral mucositis of at least 3 days (SD 6.6) at 90% power and 5% significance

Funding: "Funded by Amgen" (pharmaceutical industry)

Declarations/conflicts of interest: the majority of authors were linked to the funding pharmaceutical company in terms of employment, consulting fees, lecture fees, receipt of research funding, and ownership of equity

Data handling by review authors: N/A

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "patients were randomly assigned in a 1:1 ratio"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "patients were randomly assigned in a 1:1 ratio"

Comment: insufficient information to determine method of random sequence generation

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "placebo‐controlled, double‐blind"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "placebo‐controlled, double‐blind"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of lower grades of oral mucositis using the WHO scale, requiring the patient's assessment of pain/soreness and their ability to swallow. Higher grades have more objective elements so may not be affected by potential lack of blinding of the assessor

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 2 randomised participants (1 per group) were not included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Su 2006

Methods

Trial design: parallel (2 arms)

Location: USA

Number of centres: 1

Study duration: recruitment from January 1992 to December 1996
Trials registry number: none/unknown

Participants

Inclusion criteria: 18 years or older with histologically proven AJCC stage II to IV squamous cell carcinoma of the head and neck; undergone gross complete resection with negative pathological surgical margins and medically fit to begin RT within 8 weeks of surgery; Karnofsky performance score of 80% or more; adequate haematologic and serum metabolic laboratory indices

Exclusion criteria: nasopharyngeal cancer; concurrent active malignancy other than localised; nonmelanoma skin cancer; previous RT to head and neck; previous chemotherapy; positive serum β‐human chorionic gonadotropin in women of procreative potential; need for tube feeding at the start of RT

Cancer type: stage II to IV squamous cell carcinoma of the head and neck: hypopharynx (Group A: 11%; Group B: 5%); larynx (Group A: 16%; Group B: 23%); oral cavity (Group A: 26%; Group B: 27%); oropharynx (Group A: 21%; Group B: 32%); unknown primary (Group A: 26%; Group B: 14%)

Cancer treatment: postoperative external beam RT in once‐daily fractions of 1.8 Gy, 5 days per week, to total dose of 63 Gy at primary site and involved neck (54 Gy to regional lymphatics at risk for subclinical metastasis; spinal cord dose limited to 45 Gy)

Age at baseline (years): Group A: median 67 (interquartile range 59 to 75); Group B: median 61 (interquartile range 54 to 67)

Gender: Group A: 79% male; Group B: 73% male

Number randomised: 41 (Group A: 19; Group B: 22)

Number evaluated: 40 (Group A: 19; Group B: 21)

Interventions

Comparison: G‐CSF versus placebo

Group A: G‐CSF (3 µg/kg) by daily subcutaneous injection, 7 days per week, starting 3 days before the start of RT and continued to end of RT (total dose = dependent on duration of RT)

Group B: placebo as above

Compliance: not reported

Duration of treatment: variable and dependent on duration of RT

Outcomes

  • Oral mucositis: unknown 0 to 3 scale, where 0 = none, 1 = erythema, 2 = patchy mucositis, and 3 = confluent mucositis (assessed twice weekly by the treating radiation oncologist, maximum score reported)

  • Interruptions to cancer treatment: RT interruptions

  • Normalcy of diet (use of PEG feeding tube, as defined by 10% or higher weight loss from pre‐RT baseline)

  • Adverse events (NCI‐CTC)

  • Blood measurements (not an outcome of this review)

  • Survival (not an outcome of this review)

Notes

Sample size calculation: planned sample sample size was 66 per group to detect absolute difference of 20% in PEG tube use (from 10% in one group to 30% in the other) at 80% power and 5% significance

Funding: "This study was supported by NIH grant #CA69913" (government)

Declarations/conflicts of interest: "No benefits in any form have been or will be received from a commercial party related directly or indirectly to the subject of this article"

Data handling by review authors: although the scale for assessing oral mucositis does not completely match the 0 to 4 scales (such as WHO, etc), it was possible to dichotomise it for use in the 'any mucositis' meta‐analysis, and we also used grade 3 (confluent mucositis) as incidence of severe mucositis

Other information of note: planned to enrol 132 participants but stopped due to slow recruitment

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were randomized...by randomly permuted blocks...A randomization list was prepared by the Memorial Sloan‐Kettering Cancer Center Biostatistics Service and held by the pharmacy"

Comment: specialist centre used

Allocation concealment (selection bias)

Low risk

Quote: "A randomization list was prepared by the Memorial Sloan‐Kettering Cancer Center Biostatistics Service and held by the pharmacy. Investigators did not have access to this list, ensuring that allocation could not be predicted before registration or changed afterwards"

Comment: central/remote randomisation would have ensured concealment of the random sequence from those recruiting participants

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "All patients and treating physicians were blinded to treatment group assignment"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "All patients and treating physicians were blinded to treatment group assignment"

Comment: the treating physician was the outcome assessor

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 randomised participant was not included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Vadhan‐Raj 2010

Methods

Trial design: parallel (2 arms)

Location: University of Texas MD Anderson Cancer Center, USA

Number of centres: 1

Study duration: recruitment from December 2005 to February 2008; last follow‐up in May 2008
Trials registry number: NCT00267046 (mentioned in trial report)

Participants

Inclusion criteria: aged 15 to 65 years with sarcoma; due to start chemotherapy at the centre; Karnofsky performance score of 80% or more; adequate bone marrow, renal, and hepatic function

Exclusion criteria: history of pelvic RT; clinically significant cardiac disease; undergone surgery within the previous 2 weeks

Cancer type: sarcoma

Cancer treatment: doxorubicin (total dosage 90 mg/m²) administered by continuous IV infusion over 72 hours, and ifosfamide (total dosage 10 g/m²) administered by 3‐hour IV infusion for 4 days; participants with osteosarcoma (Group A: 2; Group B: 1) received the same dosage of doxorubicin but with intra‐arterial cisplatin (120 mg/m²); all participants received G‐CSF (pegfilgrastim) the day after chemotherapy; cycle repeated every 21 days; planned 6 cycles

Age at baseline (years): Group A: median 42 (range 17 to 63); Group B: median 39 (range 15 to 64)

Gender: Group A: 53% male; Group B: 50% male

Number randomised: 48 (Group A: 32; Group B: 16)

Number evaluated: 48 (Group A: 32; Group B: 16)

Interventions

Comparison: KGF (palifermin) versus placebo

Group A: KGF (180 µg/kg) by IV as single dose 3 days before the start of each chemotherapy cycle

Group B: placebo as above

Compliance: the proportion of participants that completed all 6 blinded cycles (i.e. they took their allocated intervention) was higher in Group A (63%) than Group B (31%)

Duration of treatment: 1 day per 3‐week cycle; planned total 6 days of intervention over 18 weeks

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed for each cycle before chemotherapy, days 10, 12, 14 and at the end of the cycle, reported as incidence of grade 2 to 4 and grade 3 to 4, reported separately for blinded phase (first 2 cycles) and then the open‐label phase) (duration of mucositis also measured but not an outcome of this review)

  • Oral pain: 0 to 10 (10 being worst) scale (assessed by questionnaire for each cycle before chemotherapy, days 10, 12, 14 and at the end of the cycle, reported for blinded phase, reported as median maximum score; unable to use data)

  • Quality of life: 1 to 7 (7 being worst) scale (assessed by questionnaire for each cycle before chemotherapy, days 10, 12, 14 and at the end of the cycle, reported for blinded phase, reported as median maximum score; unable to use data)

  • Normalcy of diet: eating and drinking assessed separately on 0 to 4 (4 being most difficult) scales (assessed by questionnaire for each cycle before chemotherapy, days 10, 12, 14 and at the end of the cycle, reported for blinded phase, reported as median maximum score; unable to use data)

  • Adverse events

  • Opioid analgesic use (reported as quantity per cycle; number of days of treatment with opioid analgesics is an outcome of this review and therefore we did not use these data)

  • Multiple patient‐reported outcomes (overall health, sleeping, dysphagia, talking, brushing teeth, throat pain, rectal soreness) (not outcomes of this review)

  • Blood and laboratory measurements (not outcomes of this review)

  • Weight loss (not an outcome of this review)

  • Survival (not an outcome of this review)

Notes

Sample size calculation: 48 participants required to detect absolute difference of 50% (from 26% in Group A to 76% in Group B) in grade 2 to 4 mucositis at 88% power and 5% significance

Funding: "Amgen provided the palifermin and partial funding for the study" (pharmaceutical industry)

Declarations/conflicts of interest: principal investigator is a member of the Amgen board

Data handling by review authors: we only report data from the first 2 cycles (blinded phase) as very few participants received placebo in the remaining cycles

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Two distinct computer‐generated randomization lists were prepared by the University of Texas M.D. Anderson Cancer Center, Department of Biostatistics, one for the 20 patients who consented to pharmacokinetic sampling and the other for the 28 patients who did not. For the pharmacokinetics cohort, the treatment allocation ratio was 4 patients receiving palifermin to 1 receiving placebo, in blocks of 5; for the other cohort, the ratio was 4 patients receiving palifermin to 3 receiving placebo, in blocks of 14"

Comment: adequate method used

Allocation concealment (selection bias)

Low risk

Quote: "The statistician provided both randomization lists to the pharmacy, so the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment. At patient enrolment, the research team notified the pharmacy, which assigned the patient the next sequential slot and treatment from the appropriate randomization list on the basis of whether he or she had consented to pharmacokinetic sampling. The pharmacy provided the research team with the blinded study medication. Upon completion of the study, pharmacy provided the statistician with the 2 randomization lists, including individual patient treatment assignments, for analysis"

Comment: pharmacy‐controlled randomisation would have ensured concealment of the random sequence from those recruiting participants

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment" and "Blinding might not have been maintained because of adverse effects of palifermin"

Comment: some personnel may not have been blinded due to increased adverse effects, although adverse effects from palifermin may have been difficult to isolate from adverse effects of cancer treatment

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "the patient and the clinical research team (who assessed outcomes) were blinded to the study treatment" and "Blinding might not have been maintained because of adverse effects of palifermin" and "patients were assessed at each cycle by both research and clinical teams, including those without direct knowledge of the protocol"

Comment: it seems unlikely that lack of blinding would affect outcomes as the higher grades of oral mucositis assessed in this study are more objective; also, some were assessed by personnel without knowledge of the protocol

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

van der Lelie 2001

Methods

Trial design: parallel (2 arms)

Location: the Netherlands

Number of centres: 1

Study duration: recruitment from May 1997 to August 1999
Trials registry number: none/unknown

Participants

Inclusion criteria: malignant disease and due to have myeloablative treatment followed by autologous or allogeneic BMT or PBSCT

Exclusion criteria: not reported

Cancer type: haematologic: lymphoma (Group A: 28%; Group B: 50%); acute leukaemia (Group A: 28%; Group B: 28%); CML (Group A: 22%; Group B: 6%); multiple myeloma (Group A: 22%; Group B: 17%)

Cancer treatment: prior to autologous or allogeneic BMT or PBSCT, participants received the following conditioning regimens:

  • BEAM (Group A: 28%; Group B: 44%): carmustine (BCNU) 300 mg/m² on day ‐6; etoposide (VP16) and cytosine arabinoside (Ara‐C) 200 mg/m² of each on days ‐5, ‐4, ‐3, ‐2; melphalan 140 mg/m² on day ‐1

  • CYTBI (Group A: 56%; Group B: 44%): cyclophosphamide 60 mg/kg on days ‐5 and ‐4; TBI 4.5 Gy on days ‐2 and ‐1

  • BUCY (Group A: 17%; Group B: 11%): busulfan 4 mg/kg on days ‐7, ‐6, ‐5, ‐4; cyclophosphamide 60 mg/kg on days ‐3 and ‐2

Lymphoma patients received BEAM; other patients received CYTBI or BUCY; nearly all participants received autologous or allogeneic PBSCT

Age at baseline (years): Group A: median 47 (range 25 to 63); Group B: median 48 (range 18 to 62)

Gender: Group A: 61% male; Group B: 39% male

Number randomised: 36 (Group A: 18; Group B: 18)

Number evaluated: 36 (Group A: 18; Group B: 18)

Interventions

Comparison: GM‐CSF versus placebo

Group A: GM‐CSF (300 µg daily dose) gel, 5 mL twice daily (early in the morning and before going to sleep) kept in the mouth for as long as possible and then swallowed; no oral intake for 1 hour afterwards; starting on day 1 (the day after the day of transplant) and continued until neutrophil recovery (typically up to 14 days after transplant)

Group B: placebo as above

All participants followed hospital's standard protocol for mouth care: toothbrushing after meals, rinsing with 0.9% saline, and if there was inflammation, 0.12% chlorhexidine rinse 6 times daily

Compliance: 8 participants (Group A: 4; Group B: 4) did not complete the study; they all cited the main reason being nausea and the taste of the gel

Duration of treatment: variable and dependent on neutrophil recovery

Outcomes

  • Oral mucositis: WHO 0 to 4 scale (assessed daily by dentists, reported in text as incidence of grade 3 to 4)

  • Oral mucositis: 8 (all 8 sites normal) to 24 (all 8 sites severely affected) oral assessment score (assessed daily by dentists, reported graphically as median score over 14 days, unable to use data)

  • Oral pain: 0 (no pain) to 100 (worst pain) VAS (assessed daily by participant, reported graphically as median score over 14 days, unable to use data)

  • Normalcy of diet: incidence of total parenteral nutrition (started when patients were unable to eat for longer than 3 days)

  • Number of days in hospital (reported as median, unable to use data)

  • Opioid analgesic use (reported as incidence; number of days of treatment with opioid analgesics is an outcome of this review and therefore we did not use these data)

  • Blood measurements (not an outcome of this review)

  • Fever (not an outcome of this review)

  • Infection (not an outcome of this review)

  • Antibiotic use (not an outcome of this review)

Notes

Sample size calculation: not reported ("GM‐CSF was supplied by the sponsor for 18 patients so that 36 patients could be included in the study. This should be enough to demonstrate a clinically significant difference")

Funding: "We thank Novartis and Schering‐Plough for supplying the GM‐CSF" (pharmaceutical industry)

Declarations/conflicts of interest: not reported

Data handling by review authors: N/A

Other information of note: GM‐CSF administration only began after the conditioning treatment had been completed, by which point oral mucositis may have already begun to develop

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "the patients were randomized to receive GM‐CSF or placebo"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "the patients were randomized to receive GM‐CSF or placebo"

Comment: insufficient information to determine method of random sequence generation

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind placebo‐controlled" and "There was no difference in taste or appearance between the placebo and the GM‐CSF"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind placebo‐controlled" and "There was no difference in taste or appearance between the placebo and the GM‐CSF"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of lower grades of oral mucositis using the WHO scale, requiring the patient's assessment of pain/soreness and their ability to swallow. Higher grades have more objective elements so may not be affected by potential lack of blinding of the assessor

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses

Selective reporting (reporting bias)

Low risk

Data for outcomes of this review were reported appropriately

Other bias

Low risk

No other sources of bias are apparent

Wu 2009

Methods

Trial design: parallel (4 arms)

Location: South Korea

Number of centres: 6

Study duration: recruitment from January to August 2007
Trials registry number: none/unknown

Participants

Inclusion criteria: 18 years or older with histological evidence of head and neck cancer; due to receive at least 5 weeks of primary RT, primary chemoradiotherapy, or postoperative RT; agreement to have complete medical history and physical examination; ECOG score of 0 to 2; white blood counts 3 x 10³/L or higher; platelet counts 100 x 10³/L or higher

Exclusion criteria: oral ulcers; herpes simplex virus; severe periodontal disease; serum creatine levels greater than 2 mg/dL; ALT/AST values greater than 200 IU/L; cytotoxic chemotherapy or RT within 3 weeks of the start of the study; systemic or topical corticosteroids within 30 days of the start of the study; participated in another clinical study within 30 days of the start of the study

Cancer type: head and neck: nasopharynx (Group A: 36%; Group B: 32%); oropharynx (Group A: 24%; Group B: 25%); oral cavity (Group A: 19%; Group B: 25%); hypopharynx (Group A: 2%; Group B: 4%); other (Group A: 19%; Group B: 14%)

Cancer treatment:

  • Radiotherapy: conventional fractionation once daily in 2 (± 0.25) Gy fractions, 5 times per week, for at least 5 weeks

  • Chemotherapy: concurrent cisplatin was allowed (just over 50% per group received this)

Age at baseline (years): Group A: median 56 (range 18 to 75); Group B: median 51 (range 18 to 77)

Gender: Group A: 69% male; Group B: 57% male

Number randomised: 113 (Group A: 85; Group B: 28)

Number evaluated: 103 (Group A: 76; Group B: 27) for incidence of moderate to severe mucositis; 94 (Group A: 70; Group B: 24) for incidence of severe mucositis

Interventions

Comparison: EGF (recombinant human) versus placebo

Group A: EGF

  • (n = 29): 10 µg daily by oral spray, applied twice daily, sprayed over the entire oral mucosa and then swallowed, no oral intake for 30 minutes afterwards; starting on first day of RT and continuing for 5 weeks

  • (n = 29): 50 µg daily as above

  • (n = 27): 100 µg daily as above

Group B: placebo as above

All participants gargled with chlorhexidine to maintain oral hygiene

Compliance: not reported

Duration of treatment: 5 weeks

Outcomes

  • Oral mucositis: RTOG 0 to 4 scale (assessed weekly by radiation oncologists, reported as incidence of grade 2 to 4 and grade 3 to 4 at week 4 or 5; unclear reporting for grade 3 to 4, unable to use data) (time to develop mucositis also assessed but not an outcome of this review)

  • WHO oral toxicity grade (assessed weekly, not clear whether this is the WHO 0 to 4 scale for oral mucositis, no data reported)

  • Interruptions to cancer treatment (incidence of 3 or more consecutive days of interruption to RT)

  • Oral pain (assessed weekly, no description of scale used, no data reported)

  • Averse events (no data reported; narrative only)

  • Opioid analgesic use (assessed weekly, not clear if duration, quantity or incidence of use was assessed, no data reported)

  • Weight (not an outcome of this review)

  • Laboratory measurements (not an outcome of this review)

Notes

Sample size calculation: allowing for 10% attrition, 26 participants per group required

Funding: "This study was supported by a grant from the National R&D Program for Cancer Central, Ministry of Health, Welfare and Family Affairs, Republic of Korea (0620270)" (government) and "The EGF and placebo treatments were supplied free of charge" (presumably pharmaceutical industry)

Declarations/conflicts of interest: not reported

Data handling by review authors: we combined the 3 EGF groups to make a single pairwise comparison against placebo; no formal statistical analysis was undertaken for the different EGF dosages

Other information of note: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "We assigned patients randomly to 4 arms"

Comment: insufficient information to determine method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Quote: "We assigned patients randomly to 4 arms"

Comment: insufficient information to determine method of random sequence generation

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled" and "Three concentrations of EGF and a placebo containing the drug delivery vehicle were manufactured, packaged, and supplied in a double‐blind fashion"

Comment: the use of a placebo should have ensured that blinding was successful

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "double‐blind, placebo‐controlled"

Comment: it is not clear who was blinded. There are subjective elements to the assessment of most grades of oral mucositis using the RTOG scale, requiring the patient's assessment of pain. Grade 4 is more objective so may not be affected by potential lack of blinding of the assessor

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall attrition was 9% (Group A: 11%; Group B: 4%). Slight differential between groups but the reasons were reported and mostly unrelated to interventions/outcomes. Unlikely to have biased the results

Selective reporting (reporting bias)

High risk

Several outcomes were assessed but not properly reported ("No secondary endpoint showed any difference between the placebo and study groups")

Other bias

Low risk

No other sources of bias are apparent

5FU = fluorouracil; AJCC = American Joint Committee on Cancer; ALL = acute lymphoblastic leukaemia; allogeneic = cells from donor; AML = acute myelogenous leukaemia; AUC = area under the curve; autologous = patients' own cells; BMT = bone marrow transplantation; CALGB = Cancer and Leukaemia Group B; cGy = centigray; CML = chronic myelogenous leukaemia; CRT = chemoradiotherapy; CT = chemotherapy; ECOG = Eastern Co‐operative Oncology Group; EGF = epidermal growth factor; EQ‐5D = European Quality Of Life Utility Scale; FACT = Functional Assessment of Cancer Therapy; FDA = US Food and Drug Administration; G‐CSF = granulocyte‐colony stimulating factor; GM‐CSF = granulocyte‐macrophage colony‐stimulating factor; GVHD = graft‐versus‐host disease; Gy = gray; HLA = human leukocyte antigen; HSCT = haematopoietic stem cell transplantation; IA = intra‐arterial; ITF = intestinal trefoil factor; ITT = intention‐to‐treat; IV = intravenous; KGF = keratinocyte growth factor; MDS = myelodysplastic syndrome; N/A = not applicable; NCI‐CTC = National Cancer Institute common toxicity criteria; NHL = non‐Hodgkin's lymphoma; NIH = National Institutes of Health; OMAS = oral mucositis assessment scale; OMDQ = oral mucositis daily questionnaire; OMWQ‐HN = Oral Mucositis Weely Questionnaire ‐ Head and Neck cancer; PBPC = peripheral blood progenitor cell; PBSCT = peripheral blood stem cell transplantation; PEG = percutaneous endoscopic gastrostomy; PP = per protocol; RT = radiotherapy; RTOG = Radiation Therapy Oncology Group; SCC = squamous cell carcinoma; SCT = stem cell transplantation; SD = standard deviation; SMD = standardised mean difference; TBI = total‐body irradiation; TGF = transforming growth factor; TPN = total parenteral nutrition; VAS = visual analogue scale; WBC = white blood cell; WHO = World Health Organization; WCCNR = Western Consortium for Cancer Nursing Research.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Antin 2002

Recombinant human interleukin‐11 versus placebo. Study stopped early due to adverse event triggering preset stopping rule ‐ data only for 10 patients in rhIL group and 3 placebo

de Koning 2007

TGF‐beta(2) versus placebo. Cross‐over study with no first period data reported (see Types of studies)

Foncuberta 2001

TGF‐beta(3) versus placebo. Participants assigned sequentially, not randomised

Gebbia 1994

G‐CSF versus thymopentin versus G‐CSF plus thymopentin versus placebo. Oral mucositis not mentioned (unknown if measured)

Gladkov 2013

G‐CSF (lipegfilgrastim) versus G‐CSF (pegfilgrastim). Oral mucositis not mentioned (unknown if measured). Insufficient information (abstract)

Gordon 1993

GM‐CSF versus no treatment. Unclear if randomised or not. Only 13 participants. Insufficient information (abstract)

Horsley 2007

KGF (palifermin) versus standard care. No random allocation

Hunter 2009

ATL‐104 versus placebo. Not RCT ‐ this study combines patients who were in cohorts with increasing doses of mouthrinse to assess safety, with an RCT

Ifrah 1999

GM‐CSF versus placebo. Primary outcome was survival/cure, with mucositis as a toxicity. Unclear if mucositis was oral or gastrointestinal

Iwase 1997

G‐CSF versus no treatment. No mention of randomisation and no description of when intervention given in relation to cancer treatment

Jones 1996

GM‐CSF versus placebo. Unclear if mucositis was oral or gastrointestinal

Karthaus 1998

G‐CSF versus placebo. Only 8 patients, 32 chemotherapy cycles and results presented assuming independent

Kubo 2016

G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim). Incidence of stomatitis reported in adverse events table
Trials registry number: JapicCTI‐111394

Lee 2016

G‐CSF (pegteograstim) versus G‐CSF (pegfilgrastim). Incidence of stomatitis reported in adverse events table
Trials registry number: NCT01328938

Legros 1997

GM‐CSF versus placebo. Unclear if mucositis was oral or gastrointestinal

Limaye 2013

AG013 versus placebo. AG013 is composed of recombinant Lactococcus lactis engineered to secrete human Trefoil Factor 1. Randomised at first but participants not developing oral mucositis in chemotherapy cycle 1 were excluded from the next phase where oral mucositis was measured, so randomisation was lost
Trials registry number: NCT00938080

Nabholtz 2002

G‐CSF (leridistim) versus G‐CSF (filgrastim). Incidence of stomatitis reported in adverse events table

NCT00360971

KGF (palifermin) versus placebo. Study terminated at 21 participants (298 planned) due to positive preliminary results from other palifermin studies

NCT00626639

KGF (palifermin) versus placebo. Study terminated at 5 participants due to departure of principal investigator and slow enrolment

Pettengell 1992

G‐CSF versus no treatment. Unclear if mucositis was oral or gastrointestinal

Ryu 2007

GM‐CSF versus placebo. Some participants (6%) had oral mucositis at baseline
Trials registry number: NCT00008398

Throuvalas 1995

GM‐CSF versus no treatment. Probably not RCT ‐ described as comparative study. Only 10 participants. Insufficient information (abstract)

Tsurusawa 2016

G‐CSF versus no treatment. Incidence of stomatitis reported in adverse events table
Trials registry number: UMIN ID: 000000675

Vitale 2014

KGF (palifermin) versus no treatment. From full text it is not RCT ‐ retrospective and allocation of KGF/no KGF based on doctor's decision

G‐CSF = granulocyte‐colony stimulating factor; GM‐CSF = granulocyte‐macrophage colony‐stimulating factor; KGF = keratinocyte growth factor; RCT = randomised controlled trial; rhIL = recombinant human interleukin; TGF = transforming growth factor.

Characteristics of studies awaiting assessment [ordered by study ID]

ACTRN12606000083594

Methods

Multicentre, double‐blind, randomised placebo‐controlled trial

Participants

18 years or older with lymphoma and due to undergo high‐dose BEAM chemotherapy and autologous stem cell transplantation as inpatient

Interventions

Whey growth factor extract at 13.5 mg/mL in sterile saline versus placebo (sterile saline)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale

  • Normalcy of diet: incidence of enteral/parenteral feeding

  • Duration of opiate analgesics

  • Quality of life: OMDQ

  • Adverse events

Notes

Funding: commercial sector/industry (TGR Biosciences, Australia)

Contact: correspondence with pharmaceutical company suggests no benefit

Antonadou 1998

Methods

Multicentre, randomised controlled trial (no placebo)

Participants

Head and neck cancer; receiving continuous course of radiotherapy for 6 to 7 weeks

Interventions

GM‐CSF (subcutaneous) versus no treatment

Outcomes

Signs and symptoms of oral mucositis: erythema, pain and dysphagia each measured on 0 to 3 scale

Notes

Abstract only (we were unable to link this abstract to a full‐text report)

Some P values suggest benefit for GM‐CSF at some time points

Quote: "GM‐CSF reduces the incidence and severity of radiation mucositis and allows the completion of the XRT treatment without protraction"

Elsaid 2001

Methods

Randomised controlled trial (no placebo)

Participants

Anaemic participants with head and neck cancer receiving radiotherapy once daily (1.8 Gy or 2 Gy) to doses of 66 Gy to 70 Gy

Interventions

Recombinant human erythropoietin versus no treatment

Outcomes

Incidence of grade 3 mucositis and dermatitis

Notes

Abstract only (we were unable to link this abstract to a full‐text report)

Higher rate of mucositis in the no‐treatment group (5.9% versus 0%)

Grzegorczyk 2006

Methods

Randomised placebo‐controlled trial

Participants

Adults (aged 19 to 68 years) undergoing haematopoietic stem cell transplantation

Interventions

G‐CSF versus placebo

Outcomes

  • Oral mucositis: WHO 0 to 4 scale

  • Oral pain: 0 to 10 VAS

  • Neutrophil counts

Notes

Translation provided insufficient information. Discrepancy between graph legends and descriptions
Unable to contact author

Koga 2015

Methods

Randomised controlled trial (no placebo)

Participants

Children with B‐cell non‐Hodgkin's lymphoma (B‐NHL) receiving chemotherapy

Interventions

G‐CSF versus no treatment

Outcomes

  • Oral mucositis (no further details)

  • Duration of hospitalisation

  • Incidence of febrile neutropenia

  • Infections

  • Time to neutrophil recovery

  • Costs

Notes

Abstract only (we were unable to link this abstract to a full‐text report)

Quote: "G‐CSF+ patients showed a positive impact on the meantime to neutrophil recovery and hospital stay, but they had no impact in incidences of febrile neutropenia, infections, stomatitis, and total cost"

NCT00293462

Methods

Double‐blind, randomised placebo‐controlled trial (1 of the 3 arms is a cross‐over)

Participants

18 years or older with head and neck cancer due to receive conventional or hyperfractionated radiotherapy or intensity‐modulated radiotherapy (IMRT) with or without concurrent chemotherapy

Interventions

GM‐CSF mouthwash versus salt and soda mouthwash versus both (cross‐over arm)

Outcomes

  • Oral mucositis: scale/s unclear

  • Quality of life: 0 to 10 scale

  • Oral pain: 0 to 10 scale

  • Functional status by Karnofsky performance status scale

Notes

Trials registry number: NCT00293462

Funding: University of California, San Francisco; National Cancer Institute (NCI)

NCT00393822

Methods

Double‐blind, randomised placebo‐controlled trial

Participants

18 years or older with resected colon cancer (American Joint Committee on Cancer Stage 2B or 3) and due to receive 5‐FU and leucovorin

Interventions

KGF versus placebo

Outcomes

  • Oral mucositis: WHO 0 to 4 scale ‐ incidence and duration of moderate to severe (grade 2 to 4)

  • Interruptions to cancer treatment

  • Mouth and throat pain

  • Adverse events

  • Survival

  • Changes in laboratory values

  • Serum anti‐KGF antibody formation

Notes

Trials registry number: NCT00393822

Funding: Amgen (pharmaceutical industry)

NCT02303197

Methods

Randomised controlled trial (no placebo)

Participants

Adults (aged 18 to 75 years) with head and neck cancer due to receive radiotherapy

Interventions

Recombinant human EGF versus Chining

Outcomes

  • Oral mucositis: RTOG 0 to 4 scale

  • Oral pain: VAS

  • Quality of life: EORTC QLQ‐H&N35

  • Weight change

  • Safety (blood/urine/kidney/liver/electrocardiogram)

Notes

Trials registry number: NCT02303197

Funding: Tianjin Medical University Cancer Institute and Hospital

NCT02313792

Methods

Double‐blind, randomised placebo‐controlled trial

Participants

16 years or older due to receive preparative cancer treatment regimen followed by autologous or allogeneic HSCT (cancer type not reported)

Interventions

KGF versus placebo

Outcomes

  • Oral mucositis: incidence and duration of severe (scale not reported)

  • Oral pain: VAS

  • Use of opioid analgesics

  • Quality of life (scale not reported)

  • Cost‐effectiveness

Notes

Trials registry number: NCT02313792

Funding: The Catholic University of Korea; Collaborator: "BLNH" (probably pharmaceutical industry)

Patte 2002

Methods

Randomised controlled trial (no placebo)

Participants

Children with non‐Hodgkin's lymphoma due to receive 2 courses of COPAD induction chemotherapy

Interventions

G‐CSF versus no treatment

Outcomes

  • Mucositis (unclear if oral or gastrointestinal): 0 to 4 scale ‐ incidence of severe (grade 3 to 4)

  • Duration of hospitalisation

  • Febrile neutropenia

  • Severe infections

  • Blood measurements

  • Use of IV antifungals or antibiotics

  • Survival

Notes

Quote: "The incidence of grade 3 and 4 mucositis was similar in both arms"
Contact: emailed corresponding author July 2017 to clarify if mucositis is oral or gastrointestinal

Schuster 2007a

Methods

Multicentre, double‐blind, randomised placebo‐controlled trial

Participants

18 years or older with multiple myeloma or lymphoma due to receive high‐dose chemotherapy (with or without TBI) followed by autologous HSCT

Interventions

Recombinant human FGF‐20 (velafermin) (3 arms with different dosages) versus placebo

Outcomes

  • Oral mucositis: WHO 0 to 4 scale ‐ incidence and duration of severe (grade 3 to 4)

  • Adverse events

Notes

Abstract only (we were unable to link this abstract to a full‐text report)

Trials registry number: NCT00104065

Schuster 2007b

Methods

Multicentre, double‐blind, randomised placebo‐controlled trial

Participants

18 years or older with multiple myeloma or lymphoma undergoing high‐dose chemotherapy (with or without TBI) followed by autologous HSCT

Interventions

Recombinant human FGF‐20 (velafermin) (3 arms with different dosages) versus placebo

Outcomes

  • Oral mucositis: WHO 0 to 4 scale ‐ incidence of severe (grade 3 to 4)

  • Adverse events

Notes

Abstract only (we were unable to link this abstract to a full‐text report)

Trials registry number: NCT00323518

Shea 2007

Methods

Randomised controlled trial (no placebo)

Participants

Aged 18 to 74 years with lymphoma, leukaemia or multiple myeloma; TBI plus high‐dose chemotherapy followed by autologous peripheral blood stem cell transplantation

Interventions

KGF (4 arms with different schedules)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale

  • Mouth and throat pain: OMDQ

  • Opioid analgesic use

  • Adverse events

Notes

Abstract only (we were unable to link this abstract to a full‐text report)

Trials registry number: NCT00109031

Spielberger 2001

Methods

Multicentre, double‐blind, randomised placebo‐controlled trial

Participants

Haematologic malignancies; eligible for TBI plus high‐dose chemotherapy followed by autologous peripheral blood stem cell transplantation; aged 12 to 65 years

Interventions

KGF (7 doses) versus KGF (4 doses + 3 doses of placebo) versus placebo (7 doses)

Outcomes

  • Oral mucositis: WHO 0 to 4 scale ‐ incidence and duration of severe (grade 3 to 4)

  • Use of opioid analgesics

  • Quality of life

  • Febrile neutropenia

  • Use of IV antifungals or antibiotics

  • Diarrhoea

Notes

Abstract only (we were unable to link this abstract to a full‐text report)

Trials registry number: NCT00004132

5FU = fluorouracil; EGF = epidermal growth factor; EORTC QLQ‐H&N35 = European Organisation for Research and Treatment of Cancer, Quality of Life Questionnaire, Head and Neck Module; G‐CSF = granulocyte‐colony stimulating factor; GM‐CSF = granulocyte‐macrophage colony‐stimulating factor; HSCT = haematopoietic stem cell transplantation; IV = intravenous; KGF = keratinocyte growth factor; NCI‐CTC = National Cancer Institute common toxicity criteria; OMDQ = oral mucositis daily questionnaire; FGF‐20 = fibroblast growth factor‐20; TBI = total‐body irradiation; VAS = visual analogue scale; WHO = World Health Organization.

Data and analyses

Open in table viewer
Comparison 1. KGF versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 KGF versus placebo, Outcome 1 Oral mucositis (any).

Comparison 1 KGF versus placebo, Outcome 1 Oral mucositis (any).

1.1 BMT/SCT after conditioning for haematological cancers

4

655

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.88, 1.05]

1.2 RT to head & neck with cisplatin

2

374

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.90, 1.00]

1.3 CT alone for mixed cancers

2

215

Risk Ratio (M‐H, Random, 95% CI)

0.71 [0.60, 0.85]

2 Oral mucositis (moderate + severe) Show forest plot

13

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 KGF versus placebo, Outcome 2 Oral mucositis (moderate + severe).

Comparison 1 KGF versus placebo, Outcome 2 Oral mucositis (moderate + severe).

2.1 BMT/SCT after conditioning for haematological cancers

6

852

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.80, 0.99]

2.2 RT to head & neck with cisplatin/5FU

3

471

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.83, 1.00]

2.3 CT alone for mixed cancers

4

344

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.45, 0.70]

3 Oral mucositis (severe) Show forest plot

12

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 KGF versus placebo, Outcome 3 Oral mucositis (severe).

Comparison 1 KGF versus placebo, Outcome 3 Oral mucositis (severe).

3.1 BMT/SCT after conditioning for haematological cancers

6

852

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.65, 1.11]

3.2 RT to head & neck with cisplatin/5FU

3

471

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.69, 0.90]

3.3 CT alone for mixed cancers

3

263

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.14, 0.65]

4 Interruptions to cancer treatment (unscheduled RT breaks of 5 or more days) Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 KGF versus placebo, Outcome 4 Interruptions to cancer treatment (unscheduled RT breaks of 5 or more days).

Comparison 1 KGF versus placebo, Outcome 4 Interruptions to cancer treatment (unscheduled RT breaks of 5 or more days).

4.1 RT to head & neck with cisplatin/5FU

3

473

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.65, 1.59]

5 Interruptions to cancer treatment (chemotherapy delays/discontinuations) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 KGF versus placebo, Outcome 5 Interruptions to cancer treatment (chemotherapy delays/discontinuations).

Comparison 1 KGF versus placebo, Outcome 5 Interruptions to cancer treatment (chemotherapy delays/discontinuations).

5.1 RT to head & neck with cisplatin

2

374

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.62, 1.47]

6 Oral pain Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.6

Comparison 1 KGF versus placebo, Outcome 6 Oral pain.

Comparison 1 KGF versus placebo, Outcome 6 Oral pain.

6.1 BMT/SCT after conditioning for haematological cancers

1

42

Mean Difference (IV, Random, 95% CI)

‐0.85 [‐3.00, 1.30]

6.2 RT to head & neck with cisplatin

2

374

Mean Difference (IV, Random, 95% CI)

‐0.12 [‐0.27, 0.02]

7 Normalcy of diet (use of supplemental nutrition) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.7

Comparison 1 KGF versus placebo, Outcome 7 Normalcy of diet (use of supplemental nutrition).

Comparison 1 KGF versus placebo, Outcome 7 Normalcy of diet (use of supplemental nutrition).

7.1 BMT/SCT after conditioning for haematological cancers

4

714

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.58, 1.34]

7.2 RT to head & neck with cisplatin/5FU

3

473

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.77, 1.37]

8 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.8

Comparison 1 KGF versus placebo, Outcome 8 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale).

Comparison 1 KGF versus placebo, Outcome 8 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale).

8.1 BMT/SCT after conditioning for haematological cancers

1

42

Mean Difference (IV, Random, 95% CI)

‐0.5 [‐1.21, 0.21]

9 Number of days in hospital Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.9

Comparison 1 KGF versus placebo, Outcome 9 Number of days in hospital.

Comparison 1 KGF versus placebo, Outcome 9 Number of days in hospital.

9.1 BMT/SCT after conditioning for haematological cancers

1

281

Mean Difference (IV, Random, 95% CI)

0.0 [‐1.64, 1.64]

10 Number of days of treatment with opioid analgesics Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.10

Comparison 1 KGF versus placebo, Outcome 10 Number of days of treatment with opioid analgesics.

Comparison 1 KGF versus placebo, Outcome 10 Number of days of treatment with opioid analgesics.

10.1 BMT/SCT after conditioning for haematological cancers

2

323

Mean Difference (IV, Random, 95% CI)

‐1.41 [‐3.33, 0.51]

Open in table viewer
Comparison 2. KGF (dose comparison)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.1

Comparison 2 KGF (dose comparison), Outcome 1 Oral mucositis (any).

Comparison 2 KGF (dose comparison), Outcome 1 Oral mucositis (any).

1.1 BMT/SCT after conditioning for haematological cancers

1

224

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.75, 1.03]

2 Oral mucositis (moderate + severe) Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 2.2

Comparison 2 KGF (dose comparison), Outcome 2 Oral mucositis (moderate + severe).

Comparison 2 KGF (dose comparison), Outcome 2 Oral mucositis (moderate + severe).

2.1 BMT/SCT after conditioning for haematological cancers

2

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 CT alone for metastatic colorectal cancer

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Oral mucositis (severe) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 2.3

Comparison 2 KGF (dose comparison), Outcome 3 Oral mucositis (severe).

Comparison 2 KGF (dose comparison), Outcome 3 Oral mucositis (severe).

3.1 BMT/SCT after conditioning for haematological cancers

2

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Oral pain (maximum score on 0 to 10 VAS) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 2.4

Comparison 2 KGF (dose comparison), Outcome 4 Oral pain (maximum score on 0 to 10 VAS).

Comparison 2 KGF (dose comparison), Outcome 4 Oral pain (maximum score on 0 to 10 VAS).

4.1 BMT/SCT after conditioning for haematological cancers

1

28

Mean Difference (IV, Random, 95% CI)

0.70 [‐1.90, 3.30]

5 Normalcy of diet (use of TPN) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.5

Comparison 2 KGF (dose comparison), Outcome 5 Normalcy of diet (use of TPN).

Comparison 2 KGF (dose comparison), Outcome 5 Normalcy of diet (use of TPN).

5.1 BMT/SCT after conditioning for haematological cancers

1

224

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.63, 1.02]

6 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 2.6

Comparison 2 KGF (dose comparison), Outcome 6 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale).

Comparison 2 KGF (dose comparison), Outcome 6 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale).

6.1 BMT/SCT after conditioning for haematological cancers

1

28

Mean Difference (IV, Random, 95% CI)

0.40 [‐0.41, 1.21]

7 Number of days in hospital Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 2.7

Comparison 2 KGF (dose comparison), Outcome 7 Number of days in hospital.

Comparison 2 KGF (dose comparison), Outcome 7 Number of days in hospital.

7.1 BMT/SCT after conditioning for haematological cancers

1

224

Mean Difference (IV, Random, 95% CI)

0.0 [‐1.78, 1.78]

8 Number of days of treatment with opioid analgesics Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.8

Comparison 2 KGF (dose comparison), Outcome 8 Number of days of treatment with opioid analgesics.

Comparison 2 KGF (dose comparison), Outcome 8 Number of days of treatment with opioid analgesics.

8.1 BMT/SCT after conditioning for haematological cancers

2

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 3. KGF versus chlorhexidine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.1

Comparison 3 KGF versus chlorhexidine, Outcome 1 Oral mucositis (any).

Comparison 3 KGF versus chlorhexidine, Outcome 1 Oral mucositis (any).

1.1 CT alone for haematological cancer

1

90

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.54, 0.85]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.2

Comparison 3 KGF versus chlorhexidine, Outcome 2 Oral mucositis (moderate + severe).

Comparison 3 KGF versus chlorhexidine, Outcome 2 Oral mucositis (moderate + severe).

2.1 CT alone for haematological cancer

1

90

Risk Ratio (M‐H, Random, 95% CI)

0.12 [0.05, 0.28]

3 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.3

Comparison 3 KGF versus chlorhexidine, Outcome 3 Oral mucositis (severe).

Comparison 3 KGF versus chlorhexidine, Outcome 3 Oral mucositis (severe).

3.1 CT alone for haematological cancer

1

90

Risk Ratio (M‐H, Random, 95% CI)

0.01 [0.00, 0.19]

Open in table viewer
Comparison 4. GM‐CSF versus placebo/no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.1

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 1 Oral mucositis (any).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 1 Oral mucositis (any).

1.1 BMT/SCT after conditioning for mixed cancers

1

90

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.80, 1.04]

1.2 RT to head & neck

1

29

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.82, 1.23]

2 Oral mucositis (moderate + severe) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.2

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 2 Oral mucositis (moderate + severe).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 2 Oral mucositis (moderate + severe).

2.1 BMT/SCT after conditioning for haematological cancers

1

109

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.79, 1.13]

2.2 RT to head & neck

1

29

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.49, 1.06]

3 Oral mucositis (severe) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.3

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 3 Oral mucositis (severe).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 3 Oral mucositis (severe).

3.1 BMT/SCT after conditioning for mixed cancers

3

235

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.33, 1.67]

3.2 RT to head & neck

1

29

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.01, 7.09]

3.3 CT alone for mixed cancers

2

65

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.05, 7.11]

4 Oral pain (maximum score on 0 to 10 VAS) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 4.4

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 4 Oral pain (maximum score on 0 to 10 VAS).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 4 Oral pain (maximum score on 0 to 10 VAS).

4.1 BMT/SCT after conditioning for mixed cancers

1

90

Mean Difference (IV, Random, 95% CI)

0.60 [‐0.85, 2.05]

5 Normalcy of diet (use of feeding tube/parenteral nutrition) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.5

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 5 Normalcy of diet (use of feeding tube/parenteral nutrition).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 5 Normalcy of diet (use of feeding tube/parenteral nutrition).

5.1 BMT/SCT after conditioning for haematological cancers

1

36

Risk Ratio (M‐H, Random, 95% CI)

1.1 [0.63, 1.91]

5.2 RT to head & neck

1

29

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.01, 7.09]

6 Number of days of treatment with opioid analgesics Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 4.6

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 6 Number of days of treatment with opioid analgesics.

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 6 Number of days of treatment with opioid analgesics.

6.1 BMT/SCT after conditioning for mixed cancers

1

90

Mean Difference (IV, Random, 95% CI)

‐1.10 [‐1.91, ‐0.29]

Open in table viewer
Comparison 5. GM‐CSF (dose comparison)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.1

Comparison 5 GM‐CSF (dose comparison), Outcome 1 Oral mucositis (severe).

Comparison 5 GM‐CSF (dose comparison), Outcome 1 Oral mucositis (severe).

1.1 CT alone for breast cancer

1

36

Risk Ratio (M‐H, Random, 95% CI)

2.75 [1.07, 7.04]

Open in table viewer
Comparison 6. GM‐CSF versus sucralfate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 6.1

Comparison 6 GM‐CSF versus sucralfate, Outcome 1 Oral mucositis (moderate + severe).

Comparison 6 GM‐CSF versus sucralfate, Outcome 1 Oral mucositis (moderate + severe).

1.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.80, 1.14]

2 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 6.2

Comparison 6 GM‐CSF versus sucralfate, Outcome 2 Oral mucositis (severe).

Comparison 6 GM‐CSF versus sucralfate, Outcome 2 Oral mucositis (severe).

2.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.24, 1.21]

3 Interruptions to cancer treatment Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 6.3

Comparison 6 GM‐CSF versus sucralfate, Outcome 3 Interruptions to cancer treatment.

Comparison 6 GM‐CSF versus sucralfate, Outcome 3 Interruptions to cancer treatment.

3.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.13 [0.01, 2.36]

4 Normalcy of diet (use of PEG tube) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 6.4

Comparison 6 GM‐CSF versus sucralfate, Outcome 4 Normalcy of diet (use of PEG tube).

Comparison 6 GM‐CSF versus sucralfate, Outcome 4 Normalcy of diet (use of PEG tube).

4.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.18 [0.01, 3.56]

Open in table viewer
Comparison 7. G‐CSF versus placebo/no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 7.1

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 1 Oral mucositis (any).

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 1 Oral mucositis (any).

1.1 RT to head & neck

2

54

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.86, 1.22]

1.2 CT alone for lung cancer

1

195

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.40, 0.87]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 7.2

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 2 Oral mucositis (moderate + severe).

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 2 Oral mucositis (moderate + severe).

2.1 CT alone for breast cancer

1

14

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.12, 0.95]

3 Oral mucositis (severe) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 7.3

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 3 Oral mucositis (severe).

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 3 Oral mucositis (severe).

3.1 RT to head & neck

2

54

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.15, 0.87]

4 Interruptions to cancer treatment (RT interruption) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 7.4

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 4 Interruptions to cancer treatment (RT interruption).

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 4 Interruptions to cancer treatment (RT interruption).

4.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.22 [0.01, 4.31]

5 Normalcy of diet (use of PEG tube) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 7.5

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 5 Normalcy of diet (use of PEG tube).

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 5 Normalcy of diet (use of PEG tube).

5.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.16 [0.01, 2.86]

Open in table viewer
Comparison 8. G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 8.1

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 1 Oral mucositis (any).

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 1 Oral mucositis (any).

1.1 BMT/SCT after conditioning for mixed cancers

1

61

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.82, 1.27]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 8.2

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 2 Oral mucositis (moderate + severe).

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 2 Oral mucositis (moderate + severe).

2.1 BMT/SCT after conditioning for mixed cancers

1

61

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.55, 1.11]

3 Normalcy of diet (use of supplemental nutrition) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 8.3

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 3 Normalcy of diet (use of supplemental nutrition).

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 3 Normalcy of diet (use of supplemental nutrition).

3.1 BMT/SCT after conditioning for mixed cancers

1

61

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.94, 1.06]

Open in table viewer
Comparison 9. EGF versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (moderate + severe) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 9.1

Comparison 9 EGF versus placebo, Outcome 1 Oral mucositis (moderate + severe).

Comparison 9 EGF versus placebo, Outcome 1 Oral mucositis (moderate + severe).

1.1 BMT/SCT after conditioning for haematological cancers

1

136

Risk Ratio (M‐H, Random, 95% CI)

1.06 [0.78, 1.43]

1.2 RT to head & neck +/‐ cisplatin

1

103

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.45, 0.99]

2 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 9.2

Comparison 9 EGF versus placebo, Outcome 2 Oral mucositis (severe).

Comparison 9 EGF versus placebo, Outcome 2 Oral mucositis (severe).

2.1 BMT/SCT after conditioning for haematological cancers

1

136

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.59, 1.80]

3 Interruptions to cancer treatment (RT breaks > 2 consecutive days) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 9.3

Comparison 9 EGF versus placebo, Outcome 3 Interruptions to cancer treatment (RT breaks > 2 consecutive days).

Comparison 9 EGF versus placebo, Outcome 3 Interruptions to cancer treatment (RT breaks > 2 consecutive days).

3.1 RT to head & neck +/‐ cisplatin

1

113

Risk Ratio (M‐H, Random, 95% CI)

4.38 [0.25, 75.44]

4 Normalcy of diet (use of supplemental nutrition) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 9.4

Comparison 9 EGF versus placebo, Outcome 4 Normalcy of diet (use of supplemental nutrition).

Comparison 9 EGF versus placebo, Outcome 4 Normalcy of diet (use of supplemental nutrition).

4.1 BMT/SCT after conditioning for haematological cancers

1

136

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.55, 1.94]

Open in table viewer
Comparison 10. ITF versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 10.1

Comparison 10 ITF versus placebo, Outcome 1 Oral mucositis (any).

Comparison 10 ITF versus placebo, Outcome 1 Oral mucositis (any).

1.1 CT alone for colorectal cancer

1

99

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.35, 0.79]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 10.2

Comparison 10 ITF versus placebo, Outcome 2 Oral mucositis (moderate + severe).

Comparison 10 ITF versus placebo, Outcome 2 Oral mucositis (moderate + severe).

2.1 CT alone for colorectal cancer

1

99

Risk Ratio (M‐H, Random, 95% CI)

0.22 [0.10, 0.48]

3 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 10.3

Comparison 10 ITF versus placebo, Outcome 3 Oral mucositis (severe).

Comparison 10 ITF versus placebo, Outcome 3 Oral mucositis (severe).

3.1 CT alone for colorectal cancer

1

99

Risk Ratio (M‐H, Random, 95% CI)

1.52 [0.06, 36.39]

Open in table viewer
Comparison 11. ITF (dose comparison)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 11.1

Comparison 11 ITF (dose comparison), Outcome 1 Oral mucositis (any).

Comparison 11 ITF (dose comparison), Outcome 1 Oral mucositis (any).

1.1 CT alone for colorectal cancer

1

66

Risk Ratio (M‐H, Random, 95% CI)

1.3 [0.67, 2.54]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 11.2

Comparison 11 ITF (dose comparison), Outcome 2 Oral mucositis (moderate + severe).

Comparison 11 ITF (dose comparison), Outcome 2 Oral mucositis (moderate + severe).

2.1 CT alone for colorectal cancer

1

66

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.18, 3.09]

3 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 11.3

Comparison 11 ITF (dose comparison), Outcome 3 Oral mucositis (severe).

Comparison 11 ITF (dose comparison), Outcome 3 Oral mucositis (severe).

3.1 CT alone for colorectal cancer

1

66

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.90]

Open in table viewer
Comparison 12. Erythropoietin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 12.1

Comparison 12 Erythropoietin versus placebo, Outcome 1 Oral mucositis (any).

Comparison 12 Erythropoietin versus placebo, Outcome 1 Oral mucositis (any).

1.1 BMT/SCT after conditioning for haematological cancers

1

80

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.21, 0.60]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 12.2

Comparison 12 Erythropoietin versus placebo, Outcome 2 Oral mucositis (moderate + severe).

Comparison 12 Erythropoietin versus placebo, Outcome 2 Oral mucositis (moderate + severe).

2.1 BMT/SCT after conditioning for haematological cancers

1

80

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.24, 0.79]

3 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 12.3

Comparison 12 Erythropoietin versus placebo, Outcome 3 Oral mucositis (severe).

Comparison 12 Erythropoietin versus placebo, Outcome 3 Oral mucositis (severe).

3.1 BMT/SCT after conditioning for haematological cancers

1

80

Risk Ratio (M‐H, Random, 95% CI)

0.4 [0.14, 1.17]

4 Number of days in hospital Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 12.4

Comparison 12 Erythropoietin versus placebo, Outcome 4 Number of days in hospital.

Comparison 12 Erythropoietin versus placebo, Outcome 4 Number of days in hospital.

4.1 BMT/SCT after conditioning for haematological cancers

1

80

Mean Difference (IV, Random, 95% CI)

‐2.95 [‐7.73, 1.83]

Study flow diagram.
 RCT = randomised controlled trial.
Figuras y tablas -
Figure 1

Study flow diagram.
RCT = randomised controlled trial.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

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

Comparison 1 KGF versus placebo, Outcome 1 Oral mucositis (any).
Figuras y tablas -
Analysis 1.1

Comparison 1 KGF versus placebo, Outcome 1 Oral mucositis (any).

Comparison 1 KGF versus placebo, Outcome 2 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 1.2

Comparison 1 KGF versus placebo, Outcome 2 Oral mucositis (moderate + severe).

Comparison 1 KGF versus placebo, Outcome 3 Oral mucositis (severe).
Figuras y tablas -
Analysis 1.3

Comparison 1 KGF versus placebo, Outcome 3 Oral mucositis (severe).

Comparison 1 KGF versus placebo, Outcome 4 Interruptions to cancer treatment (unscheduled RT breaks of 5 or more days).
Figuras y tablas -
Analysis 1.4

Comparison 1 KGF versus placebo, Outcome 4 Interruptions to cancer treatment (unscheduled RT breaks of 5 or more days).

Comparison 1 KGF versus placebo, Outcome 5 Interruptions to cancer treatment (chemotherapy delays/discontinuations).
Figuras y tablas -
Analysis 1.5

Comparison 1 KGF versus placebo, Outcome 5 Interruptions to cancer treatment (chemotherapy delays/discontinuations).

Comparison 1 KGF versus placebo, Outcome 6 Oral pain.
Figuras y tablas -
Analysis 1.6

Comparison 1 KGF versus placebo, Outcome 6 Oral pain.

Comparison 1 KGF versus placebo, Outcome 7 Normalcy of diet (use of supplemental nutrition).
Figuras y tablas -
Analysis 1.7

Comparison 1 KGF versus placebo, Outcome 7 Normalcy of diet (use of supplemental nutrition).

Comparison 1 KGF versus placebo, Outcome 8 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale).
Figuras y tablas -
Analysis 1.8

Comparison 1 KGF versus placebo, Outcome 8 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale).

Comparison 1 KGF versus placebo, Outcome 9 Number of days in hospital.
Figuras y tablas -
Analysis 1.9

Comparison 1 KGF versus placebo, Outcome 9 Number of days in hospital.

Comparison 1 KGF versus placebo, Outcome 10 Number of days of treatment with opioid analgesics.
Figuras y tablas -
Analysis 1.10

Comparison 1 KGF versus placebo, Outcome 10 Number of days of treatment with opioid analgesics.

Comparison 2 KGF (dose comparison), Outcome 1 Oral mucositis (any).
Figuras y tablas -
Analysis 2.1

Comparison 2 KGF (dose comparison), Outcome 1 Oral mucositis (any).

Comparison 2 KGF (dose comparison), Outcome 2 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 2.2

Comparison 2 KGF (dose comparison), Outcome 2 Oral mucositis (moderate + severe).

Comparison 2 KGF (dose comparison), Outcome 3 Oral mucositis (severe).
Figuras y tablas -
Analysis 2.3

Comparison 2 KGF (dose comparison), Outcome 3 Oral mucositis (severe).

Comparison 2 KGF (dose comparison), Outcome 4 Oral pain (maximum score on 0 to 10 VAS).
Figuras y tablas -
Analysis 2.4

Comparison 2 KGF (dose comparison), Outcome 4 Oral pain (maximum score on 0 to 10 VAS).

Comparison 2 KGF (dose comparison), Outcome 5 Normalcy of diet (use of TPN).
Figuras y tablas -
Analysis 2.5

Comparison 2 KGF (dose comparison), Outcome 5 Normalcy of diet (use of TPN).

Comparison 2 KGF (dose comparison), Outcome 6 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale).
Figuras y tablas -
Analysis 2.6

Comparison 2 KGF (dose comparison), Outcome 6 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale).

Comparison 2 KGF (dose comparison), Outcome 7 Number of days in hospital.
Figuras y tablas -
Analysis 2.7

Comparison 2 KGF (dose comparison), Outcome 7 Number of days in hospital.

Comparison 2 KGF (dose comparison), Outcome 8 Number of days of treatment with opioid analgesics.
Figuras y tablas -
Analysis 2.8

Comparison 2 KGF (dose comparison), Outcome 8 Number of days of treatment with opioid analgesics.

Comparison 3 KGF versus chlorhexidine, Outcome 1 Oral mucositis (any).
Figuras y tablas -
Analysis 3.1

Comparison 3 KGF versus chlorhexidine, Outcome 1 Oral mucositis (any).

Comparison 3 KGF versus chlorhexidine, Outcome 2 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 3.2

Comparison 3 KGF versus chlorhexidine, Outcome 2 Oral mucositis (moderate + severe).

Comparison 3 KGF versus chlorhexidine, Outcome 3 Oral mucositis (severe).
Figuras y tablas -
Analysis 3.3

Comparison 3 KGF versus chlorhexidine, Outcome 3 Oral mucositis (severe).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 1 Oral mucositis (any).
Figuras y tablas -
Analysis 4.1

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 1 Oral mucositis (any).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 2 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 4.2

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 2 Oral mucositis (moderate + severe).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 3 Oral mucositis (severe).
Figuras y tablas -
Analysis 4.3

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 3 Oral mucositis (severe).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 4 Oral pain (maximum score on 0 to 10 VAS).
Figuras y tablas -
Analysis 4.4

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 4 Oral pain (maximum score on 0 to 10 VAS).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 5 Normalcy of diet (use of feeding tube/parenteral nutrition).
Figuras y tablas -
Analysis 4.5

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 5 Normalcy of diet (use of feeding tube/parenteral nutrition).

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 6 Number of days of treatment with opioid analgesics.
Figuras y tablas -
Analysis 4.6

Comparison 4 GM‐CSF versus placebo/no treatment, Outcome 6 Number of days of treatment with opioid analgesics.

Comparison 5 GM‐CSF (dose comparison), Outcome 1 Oral mucositis (severe).
Figuras y tablas -
Analysis 5.1

Comparison 5 GM‐CSF (dose comparison), Outcome 1 Oral mucositis (severe).

Comparison 6 GM‐CSF versus sucralfate, Outcome 1 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 6.1

Comparison 6 GM‐CSF versus sucralfate, Outcome 1 Oral mucositis (moderate + severe).

Comparison 6 GM‐CSF versus sucralfate, Outcome 2 Oral mucositis (severe).
Figuras y tablas -
Analysis 6.2

Comparison 6 GM‐CSF versus sucralfate, Outcome 2 Oral mucositis (severe).

Comparison 6 GM‐CSF versus sucralfate, Outcome 3 Interruptions to cancer treatment.
Figuras y tablas -
Analysis 6.3

Comparison 6 GM‐CSF versus sucralfate, Outcome 3 Interruptions to cancer treatment.

Comparison 6 GM‐CSF versus sucralfate, Outcome 4 Normalcy of diet (use of PEG tube).
Figuras y tablas -
Analysis 6.4

Comparison 6 GM‐CSF versus sucralfate, Outcome 4 Normalcy of diet (use of PEG tube).

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 1 Oral mucositis (any).
Figuras y tablas -
Analysis 7.1

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 1 Oral mucositis (any).

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 2 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 7.2

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 2 Oral mucositis (moderate + severe).

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 3 Oral mucositis (severe).
Figuras y tablas -
Analysis 7.3

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 3 Oral mucositis (severe).

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 4 Interruptions to cancer treatment (RT interruption).
Figuras y tablas -
Analysis 7.4

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 4 Interruptions to cancer treatment (RT interruption).

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 5 Normalcy of diet (use of PEG tube).
Figuras y tablas -
Analysis 7.5

Comparison 7 G‐CSF versus placebo/no treatment, Outcome 5 Normalcy of diet (use of PEG tube).

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 1 Oral mucositis (any).
Figuras y tablas -
Analysis 8.1

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 1 Oral mucositis (any).

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 2 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 8.2

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 2 Oral mucositis (moderate + severe).

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 3 Normalcy of diet (use of supplemental nutrition).
Figuras y tablas -
Analysis 8.3

Comparison 8 G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim), Outcome 3 Normalcy of diet (use of supplemental nutrition).

Comparison 9 EGF versus placebo, Outcome 1 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 9.1

Comparison 9 EGF versus placebo, Outcome 1 Oral mucositis (moderate + severe).

Comparison 9 EGF versus placebo, Outcome 2 Oral mucositis (severe).
Figuras y tablas -
Analysis 9.2

Comparison 9 EGF versus placebo, Outcome 2 Oral mucositis (severe).

Comparison 9 EGF versus placebo, Outcome 3 Interruptions to cancer treatment (RT breaks > 2 consecutive days).
Figuras y tablas -
Analysis 9.3

Comparison 9 EGF versus placebo, Outcome 3 Interruptions to cancer treatment (RT breaks > 2 consecutive days).

Comparison 9 EGF versus placebo, Outcome 4 Normalcy of diet (use of supplemental nutrition).
Figuras y tablas -
Analysis 9.4

Comparison 9 EGF versus placebo, Outcome 4 Normalcy of diet (use of supplemental nutrition).

Comparison 10 ITF versus placebo, Outcome 1 Oral mucositis (any).
Figuras y tablas -
Analysis 10.1

Comparison 10 ITF versus placebo, Outcome 1 Oral mucositis (any).

Comparison 10 ITF versus placebo, Outcome 2 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 10.2

Comparison 10 ITF versus placebo, Outcome 2 Oral mucositis (moderate + severe).

Comparison 10 ITF versus placebo, Outcome 3 Oral mucositis (severe).
Figuras y tablas -
Analysis 10.3

Comparison 10 ITF versus placebo, Outcome 3 Oral mucositis (severe).

Comparison 11 ITF (dose comparison), Outcome 1 Oral mucositis (any).
Figuras y tablas -
Analysis 11.1

Comparison 11 ITF (dose comparison), Outcome 1 Oral mucositis (any).

Comparison 11 ITF (dose comparison), Outcome 2 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 11.2

Comparison 11 ITF (dose comparison), Outcome 2 Oral mucositis (moderate + severe).

Comparison 11 ITF (dose comparison), Outcome 3 Oral mucositis (severe).
Figuras y tablas -
Analysis 11.3

Comparison 11 ITF (dose comparison), Outcome 3 Oral mucositis (severe).

Comparison 12 Erythropoietin versus placebo, Outcome 1 Oral mucositis (any).
Figuras y tablas -
Analysis 12.1

Comparison 12 Erythropoietin versus placebo, Outcome 1 Oral mucositis (any).

Comparison 12 Erythropoietin versus placebo, Outcome 2 Oral mucositis (moderate + severe).
Figuras y tablas -
Analysis 12.2

Comparison 12 Erythropoietin versus placebo, Outcome 2 Oral mucositis (moderate + severe).

Comparison 12 Erythropoietin versus placebo, Outcome 3 Oral mucositis (severe).
Figuras y tablas -
Analysis 12.3

Comparison 12 Erythropoietin versus placebo, Outcome 3 Oral mucositis (severe).

Comparison 12 Erythropoietin versus placebo, Outcome 4 Number of days in hospital.
Figuras y tablas -
Analysis 12.4

Comparison 12 Erythropoietin versus placebo, Outcome 4 Number of days in hospital.

Summary of findings for the main comparison. Keratinocyte growth factor (KGF) compared to placebo for preventing oral mucositis in adults with cancer receiving treatment

KGF compared to placebo for preventing oral mucositis in adults with cancer receiving treatment

Patient or population: adults** receiving treatment for cancer (see subgroup for treatment type)
Setting: hospital
Intervention: KGF
Comparison: placebo

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Number of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with placebo

Risk with KGF

Oral mucositis (moderate + severe)

BMT/SCT after conditioning for haematological cancers

RR 0.89
(0.80 to 0.99)

852
(6 studies)

⊕⊕⊝⊝
LOW1

There might be a benefit for KGF in this population

NNTB = 11 (95% CI 6 to 112)

848 per 1000

755 per 1000
(678 to 839)

RT to head and neck with cisplatin/5FU

RR 0.91
(0.83 to 1.00)

471
(3 studies)

⊕⊕⊕⊝
MODERATE2

There is probably a benefit for KGF in this population

NNTB = 12 (95% CI 7 to ∞)

932 per 1000

848 per 1000
(773 to 932)

CT alone for mixed cancers

RR 0.56
(0.45 to 0.70)

344
(4 studies)

⊕⊕⊕⊝
MODERATE3

It is likely that there is a benefit for KGF in this population

NNTB = 4 (95% CI 3 to 6)

631 per 1000

353 per 1000
(284 to 441)

Oral mucositis (severe)

BMT/SCT after conditioning for haematological cancers

RR 0.85
(0.65 to 1.11)

852
(6 studies)

⊕⊕⊝⊝
LOW4

There might be a benefit for KGF in this population, but there is also some possibility of an increase in risk

NNTB = 10 (95% CI 5 NNTB to 14 NNTH)

677 per 1000

575 per 1000
(440 to 751)

RT to head and neck with cisplatin/5FU

RR 0.79
(0.69 to 0.90)

471
(3 studies)

⊕⊕⊕⊕
HIGH

It is very likely that there is a benefit for KGF in this population

NNTB = 7 (95% CI 5 to 15)

700 per 1000

553 per 1000
(483 to 630)

CT alone for mixed cancers

RR 0.30
(0.14 to 0.65)

263
(3 studies)

⊕⊕⊝⊝
LOW5

There might be a benefit for KGF in this population

NNTB = 10 (95% CI 8 to 19)

154 per 1000

46 per 1000
(22 to 100)

Adverse events

Adverse events that were attributed to the study drugs rather than the cancer therapy were typically oral‐related or skin‐related. Events were mostly mild to moderate with very few incidences of serious events. However, reporting was poor and inconsistent, meaning that it was not appropriate to meta‐analyse data

*The risk in the intervention group (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).
**Only 1 study in the subgroup BMT/SCT after conditioning for haematological cancers' included some children (but the median age of participants was 46 years).

***The number of people that would need to receive KGF in order to prevent 1 additional person from developing the outcome. Calculated as 1 divided by the absolute risk reduction (which is the control arm event rate minus the experimental arm event rate). NNTH means the number of people that would need to receive KGF to cause 1 additional person to develop the outcome. All decimal places have been rounded up to the nearest whole number (i.e. 6.1 = 7).

∞: infinity; 5FU: fluorouracil; BMT: bone marrow transplantation; CI: confidence interval; CT: chemotherapy; KGF: keratinocyte growth factor; NNTB: number needed to treat to benefit***; NNTH: number needed to treat to harm; RR: risk ratio; RT: radiotherapy; SCT: stem cell transplantation.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Downgraded by 1 level for inconsistency (substantial heterogeneity: I2 = 50% to 90%, P < 0.1); downgraded 1 further level for publication bias as there are 2 references in Studies awaiting classification that would be included in the conditioning/transplant subgroup, but the data are not available (NCT02313792; Spielberger 2001).
2Downgraded by 1 level for inconsistency (substantial heterogeneity: I2 = 50% to 90%, P < 0.1).
3Downgraded by 1 level for publication bias as there is 1 reference in Studies awaiting classification that would be included in the chemotherapy alone subgroup, but the data are not available (NCT00393822).
4Downgraded by 1 level for inconsistency (substantial heterogeneity: I2 = 50% to 90%, P < 0.1); downgraded 1 further level for publication bias as there are 2 references in Studies awaiting classification that would be included in the conditioning/transplant subgroup, but the data are not available (NCT02313792; Spielberger 2001); we did not downgrade for imprecision because, despite the confidence interval including a small chance of an increase in risk, it is a fairly narrow interval and a rating of 'very low quality' would seem an overly harsh rating for this body of evidence.
5Downgraded by 1 level for imprecision (wide confidence interval, small sample size and low event rate); downgraded 1 further level for publication bias as there is 1 reference in Studies awaiting classification that would be included in the chemotherapy alone subgroup, but the data are not available (NCT00393822).

Figuras y tablas -
Summary of findings for the main comparison. Keratinocyte growth factor (KGF) compared to placebo for preventing oral mucositis in adults with cancer receiving treatment
Summary of findings 2. Granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) compared to placebo/no treatment for preventing oral mucositis in adults with cancer receiving treatment

GM‐CSF compared to placebo/no treatment for preventing oral mucositis in adults with cancer receiving treatment

Patient or population: adults** receiving treatment for cancer (see subgroup for treatment type)
Setting: hospital
Intervention: GM‐CSF
Comparison: placebo/no treatment

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Number of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with placebo/no treatment

Risk with GM‐CSF

Oral mucositis (moderate + severe)

BMT/SCT after conditioning for haematological cancers

RR 0.94
(0.79 to 1.13)

109
(1 study)

⊕⊝⊝⊝
VERY LOW1

There is insufficient evidence to determine a benefit for GM‐CSF in this population

NNTB = 20 (95% CI 6 NNTB to 10 NNTH)

839 per 1000

789 per 1000
(663 to 948)

RT to head and neck

RR 0.72
(0.49 to 1.06)

29
(1 study)

⊕⊝⊝⊝
VERY LOW2

There is insufficient evidence to determine a benefit for GM‐CSF in this population

NNTB = 4 (95% CI 3 NNTB to 14 NNTH)

929 per 1000

669 per 1000
(455 to 984)

Oral mucositis (severe)

BMT/SCT after conditioning for mixed cancers

RR 0.74
(0.33 to 1.67)

235
(3 studies)

⊕⊕⊝⊝
LOW3

There is insufficient evidence to determine a benefit for GM‐CSF in this population

NNTB = 12 (95% CI 5 NNTB to 5 NNTH)

347 per 1000

257 per 1000
(115 to 580)

RT to head and neck

RR 0.31
(0.01 to 7.09)

29
(1 study)

⊕⊝⊝⊝
VERY LOW4

There is insufficient evidence to determine a benefit for GM‐CSF in this population

NNTB = 21 (95% CI 15 NNTB to 3 NNTH)

71 per 1000

22 per 1000
(1 to 506)

CT alone for mixed cancers

RR 0.59
(0.05 to 7.11)

65
(2 studies)

⊕⊝⊝⊝
VERY LOW5

There is insufficient evidence to determine a benefit for GM‐CSF in this population

NNTB = 5 (95% CI 3 NNTB to 2 NNTH)

500 per 1000

295 per 1000
(25 to 1000)

Adverse events

Adverse events that were attributed to the study drugs rather than the cancer therapy were typically bone pain, nausea, fever and headache. Events were not reported as being serious. Some studies did not report adverse events and 1 even reported that there were none. However, reporting was poor and inconsistent, meaning that it was not appropriate to meta‐analyse data

*The risk in the intervention group (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).
**There were no studies conducted on children.

***The number of people that would need to receive GM‐CSF in order to prevent 1 additional person from developing the outcome. Calculated as 1 divided by the absolute risk reduction (which is the control arm event rate minus the experimental arm event rate). NNTH means the number of people that would need to receive GM‐CSF to cause 1 additional person to develop the outcome. All decimal places have been rounded up to the nearest whole number (i.e. 6.1 = 7).

BMT: bone marrow transplantation; CI: confidence interval; CT: chemotherapy; GM‐CSF: granulocyte‐macrophage colony‐stimulating factor; NNTB: number needed to treat to benefit***; NNTH: number needed to treat to harm; RR: risk ratio; RT: radiotherapy; SCT: stem cell transplantation.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Downgraded by 2 levels for imprecision (single study with a small sample size and the confidence interval includes a possible increase in risk that is of a similar magnitude to the possible reduction in risk); downgraded 1 further level for indirectness (single study so not widely generalisable).
2Downgraded by 2 levels for imprecision (wide confidence interval and very small sample size); downgraded by 1 further level for high risk of performance bias; downgraded by 1 further level for indirectness (single study so not widely generalisable); downgraded by 1 further level for publication bias as there are 2 references in Studies awaiting classification that would be included in the RT to head and neck subgroup, but the data are not currently available (Antonadou 1998; NCT00293462).
3Downgraded by 2 levels for imprecision (small sample size and the confidence interval includes a possible increase in risk that is of a similar magnitude to the possible reduction in risk); downgraded by 1 further level for inconsistency (substantial heterogeneity: I2 = 50% to 90%, P < 0.1).
4Downgraded by 2 levels for imprecision (extremely wide confidence interval incorporating both very large increase and reduction in risk, very small sample size and very low event rate); downgraded by 1 further level for high risk of performance bias; downgraded by 1 further level for indirectness (single study so not widely generalisable); downgraded by 1 further level for publication bias as there are 2 references in Studies awaiting classification that would be included in the RT to head and neck subgroup, but the data are not currently available (Antonadou 1998; NCT00293462).
5Downgraded by 2 levels for imprecision (extremely wide confidence interval incorporating both very large increase and reduction in risk and very small sample size); downgraded by 1 further level for high risk of performance bias; downgraded by 1 further level for inconsistency (substantial heterogeneity: I2 = 50% to 90%, P < 0.1).

Figuras y tablas -
Summary of findings 2. Granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) compared to placebo/no treatment for preventing oral mucositis in adults with cancer receiving treatment
Summary of findings 3. Granulocyte‐colony stimulating factor (G‐CSF) compared to placebo/no treatment for preventing oral mucositis in adults with cancer receiving treatment

G‐CSF compared to placebo/no treatment for preventing oral mucositis in patients with cancer receiving treatment

Patient or population: adults** receiving treatment for cancer (see subgroup for treatment type)
Setting: hospital
Intervention: G‐CSF
Comparison: placebo/no treatment

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Number of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with placebo/no treatment

Risk with G‐CSF

Oral mucositis (moderate + severe)

CT alone for breast cancer

RR 0.33
(0.12 to 0.95)

14
(1 study)

⊕⊝⊝⊝
VERY LOW1

There is very weak evidence that there might be a benefit for G‐CSF in this population

NNTB = 2 (95% CI 2 to 20)

1000 per 1000

330 per 1000
(120 to 950)

Oral mucositis (severe)

RT to head and neck

RR 0.37
(0.15 to 0.87)

54
(2 studies)

⊕⊕⊝⊝
LOW2

There is weak evidence that there might be a benefit for G‐CSF in this population

NNTB = 3 (95% CI 3 to 15)

519 per 1000

192 per 1000
(78 to 451)

Adverse events

There was limited evidence of adverse events for G‐CSF. 2 of the 6 studies did not report adverse events. There were low rates of mild to moderate events, the most common of which appeared to be bone pain. However, reporting was poor and inconsistent, meaning that it was not appropriate to meta‐analyse data

*The risk in the intervention group (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).
**There were no studies conducted on children.

***The number of people that would need to receive G‐CSF in order to prevent 1 additional person from developing the outcome. Calculated as 1 divided by the absolute risk reduction (which is the control arm event rate minus the experimental arm event rate). NNTH means the number of people that would need to receive G‐CSF to cause 1 additional person to develop the outcome. All decimal places have been rounded up to the nearest whole number (i.e. 6.1 = 7).

CI: confidence interval; CT: chemotherapy; G‐CSF: granulocyte‐colony stimulating factor; NNTB: number needed to treat to benefit***; NNTH: number needed to treat to harm; RR: risk ratio; RT: radiotherapy.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Downgraded by 2 levels for imprecision (wide confidence interval and very small sample size); downgraded by 1 further level for high risk of performance bias; downgraded by 1 further level for indirectness (single study so not widely generalisable).
2Downgraded by 2 levels for imprecision (wide confidence interval and very small sample size).

Figuras y tablas -
Summary of findings 3. Granulocyte‐colony stimulating factor (G‐CSF) compared to placebo/no treatment for preventing oral mucositis in adults with cancer receiving treatment
Table 1. Adverse events: KGF

Study ID

Adverse events results

Blazar 2006

  • AEs with incidence ≥ 10% greater in KGF group: higher rate of skin rash in KGF group (65/69 versus 21/31; RR 1.39, 95% CI 1.08 to 1.79; P = 0.01). Insufficient evidence of a difference in edema, infection, or local pain

  • Grade 3 to 4 (WHO and NCI‐CTC 0 to 4 toxicities scales) AEs with higher incidence in KGF group: insufficient evidence of a difference in skin reactions, diarrhoea, local pain, or cardiac events

Blijlevens 2013

  • KGF‐related AE (NCI‐CTC): higher rate in KGF group (141/220 versus 17/57; RR 2.15, 95% CI 1.43 to 3.24; P = 0.0003)

  • KGF‐related serious AE (no definition of 'serious' given) (NCI‐CTC): insufficient evidence of a difference (4/220 versus 0/57; P = 0.56)

  • KGF‐related severe AE (NCI‐CTC grade 3, 4 or 5) (grade 5 = death): insufficient evidence of a difference (23/220 versus 0/57; P = 0.08)

Bradstock 2014

Insufficient evidence of a difference in infection or Grade 3 to 4 (NCI‐CTC 0 to 4 toxicity scale) skin rash/desquamation

Brizel 2008

The study authors state that most adverse events were considered to be caused by the cancer treatment or the underlying cancer itself and not related to study treatment. 2 participants in the palifermin group had serious adverse events considered to be related to the intervention: 1 had increased sputum production; the other had dehydration, dysphagia, pain (including abdominal), pancreatitis, and subsequently had schistosomiasis

Fink 2011

Total of 28 side effects in palifermin group occurring in 11 of 22 patients (50%) who received at least 4 of the 6 doses. Most frequent (90.9%) were cases of erythema or exanthema, often associated with itching (54.5%). Often (54.5%) a swelling of the oral mucosa including the tongue occurred. In 4 out of 6 patients, this was accompanied by taste disturbance. The severity of side effects were classified as mild to moderate. The CTC Grade 3 occurred only once in the form of a strong heat sensation

In 1 of the 11 cases, there was premature discontinuation of palifermin due to severe facial swelling with eyelid and laryngeal pain as well as painful swelling of the hands following the second injection

Freytes 2004

25 different adverse events were reported and were mostly not KGF‐related. There was insufficient evidence of a difference for diarrhoea, abdominal pain, infection or rash

Gholizadeh 2016

"..two patients reported knee joint pain, skin rash was observed in one patient, two patients had abnormal taste, and one showed lingual mucosal thickening" (control group was chlorhexidine mouthrinse. The authors do not report the events by treatment group)

Henke 2011

  • "Initially, patients were allocated to three arms: palifermin (180 g/kg/wk) throughout radiochemotherapy (ie, for at least seven doses); palifermin (180 g/kg/wk) for four doses and then placebo throughout the remainder of radiochemotherapy; or placebo throughout radiochemotherapy. However, after one serious adverse event of respiratory insufficiency was reported in one of the first 10 patients, the data monitoring committee concluded that the study should be restarted with a lower palifermin dose (120 g/kg/wk)"

  • "Most patients (97%) experienced at least one adverse event......One serious adverse event (febrile neutropenia) considered related to study drug was reported for one patient in the palifermin arm"

Jagasia 2012

KGF‐related AEs with incidence ≥ 5% in KGF group: higher rate of gastrointestinal disorders in KGF group (18/78 versus 2/73; RR 8.42, 95% CI 2.02 to 35.04; P = 0.003). Insufficient evidence of a difference in any AE, tongue coating, tongue disorder, skin and subcutaneous tissue disorders, rash, pruritus, or erythema

Le 2011

"Study drug–related AEs were reported for 35% of palifermin and 11% of placebo patients. The most frequent study drug–related AEs (palifermin, placebo) were rash (9%, 2%), flushing (5%, 0%), dysgeusia (5%, 1%), nausea (4%, 1%),and vomiting (3%, 1%). None of these events led to study withdrawal. Serious AEs considered related to study treatment were reported for five palifermin patients (5%; one each with necrotic pancreatitis, hypersensitivity, tracheostomy malfunction, peritoneal carcinoma, and convulsion) and two placebo patients (2%; one each with hepatitis/hepatic enzyme increase and cryptogenic organizing pneumonia)"

Lucchese 2016a

"The administration of palifermin was generally safe and without considerable complications. The only adverse reactions were rashes (lasting for 48–72 hours) localized to the face, upper neck and shoulders, erythema, and altered taste (consistent with the pharmacologic action of palifermin of oral epithelium and skin), most of which were of NCI grade 1 or 2 severity"

Lucchese 2016b

"The administration of palifermin was mostly safe and without substantial complications. The mean duration of the OM and the number of adverse event was significant less in the palifermin group (Tables II, III, Figure 1). The main adverse episodes were erythema, cutaneous rashes and altered taste and three of the patients in the palifermin group showed a light thickness of the tongue, mouth and palate"

Meropol 2003

"Although the predefined frequency of DLTs attributable to KGF was not reached with KGF doses between 1 and 80 µg/kg/d, there were three adverse reactions involving the skin that required discontinuation of KGF in the 18 patients treated with 60 or 80 µg/kg (Table 5). Overall, skin and oral adverse events (rash, flushing, pruritis, edema, hypoesthesia, paresthesia, tongue disorder [thickening], and alteration in taste sensation) attributed to KGF occurred in 13 of 18 patients treated with 60 and 80 µg/kg of KGF (eight patients, grade 1; four patients, grade 2; and one patient, grade 3) and in three of 11 patients treated with 40 µg/kg (all grade 1). These events were reported in 16 of 39 patients (41%) dosed with KGF at > 20 µg/kg/d, whereas these symptoms were reported in only two of 21 subjects (10%) treated with placebo. The skin and oral toxicities associated with KGF were generally mild to moderate in severity, with onset approximately 36 hours after the first dose of KGF and resolution 7 to 10 days thereafter"

Rosen 2006

  • "As expected based on the pharmacologic activity of palifermin, oral‐related AEs were reported more frequently in palifermin than in patients receiving placebo (Table 3). During cycle 1, 50% of patients receiving palifermin experienced an oral‐related AE, compared with 33% of patients receiving placebo (P = 0.13). Similarly, 56% of patients receiving palifermin during the second chemotherapy cycle had at least one oral‐related AE, compared with 38% of patients receiving placebo (P = 0.26). The overall incidences of skin‐related AEs, reported as a palifermin‐related AE in other clinical settings, were comparable between the two treatment groups (Table 3). During cycle 1, skin‐related AEs were 56% in the placebo group versus 43% in patients receiving palifermin. During cycle 2, these incidences between the two groups were comparable (palifermin, 52%; placebo, 50%)"

  • There were no serious KGF‐related AEs in either group and either cycle

Spielberger 2004

"The incidence, frequency, and severity of adverse events were similar in the two groups, and most were attributable to the underlying cancer, cytotoxic chemotherapy, or total‐body irradiation. Those that occurred with an incidence that was at least 5 percentage points higher in the palifermin group than in the placebo group are listed in Table 3. Most of these adverse events were consistent with the pharmacologic action of palifermin on skin and oral epithelium (e.g., rash, pruritus, erythema, paresthesia, mouth and tongue disorders, and taste alteration). All these events were mild to moderate in severity, transient (occurring approximately three days after the third dose of palifermin and lasting approximately three days), and not a cause for the discontinuation of study drug. Serious adverse events considered to be related to treatment occurred in one palifermin recipient (rash) and one placebo recipient (hypotension)"

Vadhan‐Raj 2010

  • "Many patients who received palifermin sensed thickening of the oral mucosa and tongue" (first 2 blinded cycles: 72% versus 31%, P = 0.007)

  • "Treatment with palifermin was well tolerated. Table 3 shows the common adverse effects that occurred during the first 2 blinded cycles, which included symptoms of thickness of oral mucosa, tongue, and lips (Figure 4); altered taste; flushing; warm sensation; and increased saliva. These adverse effects were mild to moderate and transient in nature. Similar side effects were observed during later cycles...but they did not worsen in severity"

AE = adverse event; CI = confidence interval; KGF = keratinocyte growth factor; NCI‐CTC = National Cancer Institute common toxicity criteria; RR = risk ratio; WHO = World Health Organization.

Figuras y tablas -
Table 1. Adverse events: KGF
Table 2. Adverse events: GM‐CSF

Study ID

Adverse events results

Cartee 1995

2 participants (group allocation not reported) withdrew by day 3 due to intolerance to their mouthwash (dry mouth); 1 participant receiving GM‐CSF (1 µg/mL) had mouthwash withdrawn by day 3 due to possible allergic reaction (sensation of fullness in the posterior pharyngeal area) but resolved within 4 hours (the participant was withdrawn but appears to have been included in the analysis)

Chi 1995

"One patient had fever and chills, and two patients had general malaise and headache during GM‐CSF treatment. No patient had evidence of fluid retention after GM‐CSF"

Dazzi 2003

Not reported

Makkonen 2000

(Sucralfate given to both groups) "Only 2 of the 20 patients treated with GM‐CSF and sucralfate did not experience any side effects related to the drugs, but most side effects were mild (WHO Grade 1 or 2). The most common side effects were local skin reactions, fever, bone pain, and mild nausea...In the control group only 1 patient complained of nausea possibly related to the use of sucralfate, and another patient interrupted sucralfate treatment because of the same reason"

McAleese 2006

"12 patients who received GM‐CSF had elevated white cell counts (WCC). The range of maximal WCC was 7.2–30.5 (median 19.7). All WCC had returned to normal within 3 weeks of completing injections (median 2 weeks). Three patients developed influenza‐like symptoms with the GM‐CSF and in one patient the injections were stopped because of this symptom. One patient developed an erythematous rash at his injection sites after completing his course of 14 injections (Figure 3). He had a past history of allergy to radiographic contrast medium"

Nemunaitis 1995

  • "The incidences of grades III or IV toxicities between rhGM‐CSF or placebo occurring with a > 10% frequency included anorexia (38% vs. 36%), nausea (26% vs. 29%), diarrhea (19% vs. 7%), stomatitis (19% vs. 14%) and hypertension (13% vs. 20%)"

  • "The following events were reported with higher frequency in the rhGM‐CSF group compared with placebo: diarrhea (81% vs. 66%), bone pain (21% vs. 5%), abdominal pain (38% vs. 23%), vomiting (70% vs. 57%), pharyngitis (23% vs. 13%), pruritis (23% vs. 13%) and occular hemorrhage (11% vs. 0%)"

  • "Placebo‐treated patients had higher occurrence of unspecified pain (36% vs. 17%), back pain (18% vs. 9%), peripheral edema (21% vs. 15%), hematuria (21% vs. 9%) and pneumonia (7% vs. 0%)"

Saarilahti 2002

(Comparator was sucralfate) "Both mouthwashes were well tolerated, and none of the patients reported any adverse effects related to the mouthwashes. Adverse effects commonly associated with subcutaneous GM‐CSF administration, such as nausea, vomiting, bone pain, headaches, and fever, were not observed"

van der Lelie 2001

Not reported

GM‐CSF = granulocyte‐macrophage colony‐stimulating factor.

Figuras y tablas -
Table 2. Adverse events: GM‐CSF
Table 3. G‐CSF versus placebo

Study ID

Population

Outcome

GM‐CSF

Placebo

Result

Linch 1993

BMT/SCT after conditioning for haematological cancers

Oral mucositis: no scale described

No data

No data

"There was no difference in the frequency of stomatitis (defined as a sore, infected or ulcerated mouth, lips or pharynx), the incidence being between 29 and 33% in all groups"

BMT = bone marrow transplantation; G‐CSF: granulocyte‐colony stimulating factor; SCT = stem cell transplantation.

Figuras y tablas -
Table 3. G‐CSF versus placebo
Table 4. Adverse events: G‐CSF

Study ID

Adverse events results

Cesaro 2013

"Both pegfilgrastim and filgrastim were well tolerated and no significant adverse effects were associated with their use" (G‐CSF versus G‐CSF)

Crawford 1999

Approximately 20% of participants receiving G‐CSF experienced mild to moderate skeletal pain which was resolved by using oral analgesics; 6% of participants in both groups reported mild generalised rash/itching; 3 participants experienced an event thought to be G‐CSF‐related and which caused them to request withdrawal from the study: abdominal pain, diffuse aches and pains, and a flare‐up of pre‐existing eczema

Katano 1995

Not reported

Linch 1993

"There was no difference in the overall frequency of adverse clinical or laboratory events between the groups or in the frequency of adverse events thought by the clinicians to be possibly or probably due to study medication"

Schneider 1999

Not reported

Su 2006

"In general, toxicities typical of postoperative RT to the head and neck were observed. Additional toxicities attributable to G‐CSF and/or daily injections were as follows: elevated WBC requiring G‐CSF dose reduction by prospectively planned guidelines occurred in nine patients in the GCSF arm; grade 2–3 bone pain was observed in two patients in the G‐CSF arm; three patients refused injection (2 G‐CSF, 1 placebo)"

G‐CSF = granulocyte‐colony stimulating factor.

Figuras y tablas -
Table 4. Adverse events: G‐CSF
Table 5. Adverse events: EGF

Study ID

Adverse events results

Kim 2017

"Adverse events were similar in both groups (Table 3). The most common adverse event in the rhEGF group was nausea (n = 7, 10.4%). The incidence of other adverse events including oral pain, dry mouth, and taste alteration was low. All the adverse events were mild and transient. No grade 3 or 4 adverse events were noted during the study period" (there were no differences between groups in any adverse event)

Wu 2009

"The frequency of minor and serious adverse events was similar in all groups. Most adverse events were related to primary disease status and treatment modalities"

EGF = epidermal growth factor.

Figuras y tablas -
Table 5. Adverse events: EGF
Table 6. Adverse events: ITF

Study ID

Adverse events results

Peterson 2009

"Only a minority of patients (six [6.1%] of 99 patients) reported mild to moderate treatment‐emergent adverse events on the study. The symptoms included abdominal pain, diarrhea, oral pain, headache, and hypertension (Table 2). Of these, four were considered related to study drug: one (3%) was in the placebo group, two (6%) were in the low‐dose rhITF group, and one (3%) was in the high‐dose rhITF group. The events were isolated and resolved spontaneously without sequelae"

ITF = intestinal trefoil factor.

Figuras y tablas -
Table 6. Adverse events: ITF
Table 7. TGF‐beta(2) versus placebo

Study ID

Population

Outcome

TGF‐beta(2)

Placebo

Result

Antoun 2009

CT alone for colorectal cancer

Oral mucositis (WHO 0 to 4 scale): any oral mucositis

0/9

2/4

RR 0.10 (95% CI 0.01 to 1.71); P = 0.11

CI = confidence interval; CT = chemotherapy; RR = risk ratio; TGF = transforming growth factor; WHO = World Health Organization.

Figuras y tablas -
Table 7. TGF‐beta(2) versus placebo
Comparison 1. KGF versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 BMT/SCT after conditioning for haematological cancers

4

655

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.88, 1.05]

1.2 RT to head & neck with cisplatin

2

374

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.90, 1.00]

1.3 CT alone for mixed cancers

2

215

Risk Ratio (M‐H, Random, 95% CI)

0.71 [0.60, 0.85]

2 Oral mucositis (moderate + severe) Show forest plot

13

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 BMT/SCT after conditioning for haematological cancers

6

852

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.80, 0.99]

2.2 RT to head & neck with cisplatin/5FU

3

471

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.83, 1.00]

2.3 CT alone for mixed cancers

4

344

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.45, 0.70]

3 Oral mucositis (severe) Show forest plot

12

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 BMT/SCT after conditioning for haematological cancers

6

852

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.65, 1.11]

3.2 RT to head & neck with cisplatin/5FU

3

471

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.69, 0.90]

3.3 CT alone for mixed cancers

3

263

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.14, 0.65]

4 Interruptions to cancer treatment (unscheduled RT breaks of 5 or more days) Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 RT to head & neck with cisplatin/5FU

3

473

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.65, 1.59]

5 Interruptions to cancer treatment (chemotherapy delays/discontinuations) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 RT to head & neck with cisplatin

2

374

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.62, 1.47]

6 Oral pain Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

6.1 BMT/SCT after conditioning for haematological cancers

1

42

Mean Difference (IV, Random, 95% CI)

‐0.85 [‐3.00, 1.30]

6.2 RT to head & neck with cisplatin

2

374

Mean Difference (IV, Random, 95% CI)

‐0.12 [‐0.27, 0.02]

7 Normalcy of diet (use of supplemental nutrition) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

7.1 BMT/SCT after conditioning for haematological cancers

4

714

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.58, 1.34]

7.2 RT to head & neck with cisplatin/5FU

3

473

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.77, 1.37]

8 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

8.1 BMT/SCT after conditioning for haematological cancers

1

42

Mean Difference (IV, Random, 95% CI)

‐0.5 [‐1.21, 0.21]

9 Number of days in hospital Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

9.1 BMT/SCT after conditioning for haematological cancers

1

281

Mean Difference (IV, Random, 95% CI)

0.0 [‐1.64, 1.64]

10 Number of days of treatment with opioid analgesics Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

10.1 BMT/SCT after conditioning for haematological cancers

2

323

Mean Difference (IV, Random, 95% CI)

‐1.41 [‐3.33, 0.51]

Figuras y tablas -
Comparison 1. KGF versus placebo
Comparison 2. KGF (dose comparison)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 BMT/SCT after conditioning for haematological cancers

1

224

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.75, 1.03]

2 Oral mucositis (moderate + severe) Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 BMT/SCT after conditioning for haematological cancers

2

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 CT alone for metastatic colorectal cancer

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Oral mucositis (severe) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 BMT/SCT after conditioning for haematological cancers

2

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Oral pain (maximum score on 0 to 10 VAS) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 BMT/SCT after conditioning for haematological cancers

1

28

Mean Difference (IV, Random, 95% CI)

0.70 [‐1.90, 3.30]

5 Normalcy of diet (use of TPN) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 BMT/SCT after conditioning for haematological cancers

1

224

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.63, 1.02]

6 Normalcy of diet (worst ability to eat score ‐ 1 to 4 scale) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

6.1 BMT/SCT after conditioning for haematological cancers

1

28

Mean Difference (IV, Random, 95% CI)

0.40 [‐0.41, 1.21]

7 Number of days in hospital Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

7.1 BMT/SCT after conditioning for haematological cancers

1

224

Mean Difference (IV, Random, 95% CI)

0.0 [‐1.78, 1.78]

8 Number of days of treatment with opioid analgesics Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Totals not selected

8.1 BMT/SCT after conditioning for haematological cancers

2

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 2. KGF (dose comparison)
Comparison 3. KGF versus chlorhexidine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 CT alone for haematological cancer

1

90

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.54, 0.85]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 CT alone for haematological cancer

1

90

Risk Ratio (M‐H, Random, 95% CI)

0.12 [0.05, 0.28]

3 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 CT alone for haematological cancer

1

90

Risk Ratio (M‐H, Random, 95% CI)

0.01 [0.00, 0.19]

Figuras y tablas -
Comparison 3. KGF versus chlorhexidine
Comparison 4. GM‐CSF versus placebo/no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 BMT/SCT after conditioning for mixed cancers

1

90

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.80, 1.04]

1.2 RT to head & neck

1

29

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.82, 1.23]

2 Oral mucositis (moderate + severe) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 BMT/SCT after conditioning for haematological cancers

1

109

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.79, 1.13]

2.2 RT to head & neck

1

29

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.49, 1.06]

3 Oral mucositis (severe) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 BMT/SCT after conditioning for mixed cancers

3

235

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.33, 1.67]

3.2 RT to head & neck

1

29

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.01, 7.09]

3.3 CT alone for mixed cancers

2

65

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.05, 7.11]

4 Oral pain (maximum score on 0 to 10 VAS) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 BMT/SCT after conditioning for mixed cancers

1

90

Mean Difference (IV, Random, 95% CI)

0.60 [‐0.85, 2.05]

5 Normalcy of diet (use of feeding tube/parenteral nutrition) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 BMT/SCT after conditioning for haematological cancers

1

36

Risk Ratio (M‐H, Random, 95% CI)

1.1 [0.63, 1.91]

5.2 RT to head & neck

1

29

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.01, 7.09]

6 Number of days of treatment with opioid analgesics Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

6.1 BMT/SCT after conditioning for mixed cancers

1

90

Mean Difference (IV, Random, 95% CI)

‐1.10 [‐1.91, ‐0.29]

Figuras y tablas -
Comparison 4. GM‐CSF versus placebo/no treatment
Comparison 5. GM‐CSF (dose comparison)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 CT alone for breast cancer

1

36

Risk Ratio (M‐H, Random, 95% CI)

2.75 [1.07, 7.04]

Figuras y tablas -
Comparison 5. GM‐CSF (dose comparison)
Comparison 6. GM‐CSF versus sucralfate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.80, 1.14]

2 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.24, 1.21]

3 Interruptions to cancer treatment Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.13 [0.01, 2.36]

4 Normalcy of diet (use of PEG tube) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.18 [0.01, 3.56]

Figuras y tablas -
Comparison 6. GM‐CSF versus sucralfate
Comparison 7. G‐CSF versus placebo/no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 RT to head & neck

2

54

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.86, 1.22]

1.2 CT alone for lung cancer

1

195

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.40, 0.87]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 CT alone for breast cancer

1

14

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.12, 0.95]

3 Oral mucositis (severe) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 RT to head & neck

2

54

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.15, 0.87]

4 Interruptions to cancer treatment (RT interruption) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.22 [0.01, 4.31]

5 Normalcy of diet (use of PEG tube) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 RT to head & neck

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.16 [0.01, 2.86]

Figuras y tablas -
Comparison 7. G‐CSF versus placebo/no treatment
Comparison 8. G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 BMT/SCT after conditioning for mixed cancers

1

61

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.82, 1.27]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 BMT/SCT after conditioning for mixed cancers

1

61

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.55, 1.11]

3 Normalcy of diet (use of supplemental nutrition) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 BMT/SCT after conditioning for mixed cancers

1

61

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.94, 1.06]

Figuras y tablas -
Comparison 8. G‐CSF (pegfilgrastim) versus G‐CSF (filgrastim)
Comparison 9. EGF versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (moderate + severe) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 BMT/SCT after conditioning for haematological cancers

1

136

Risk Ratio (M‐H, Random, 95% CI)

1.06 [0.78, 1.43]

1.2 RT to head & neck +/‐ cisplatin

1

103

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.45, 0.99]

2 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 BMT/SCT after conditioning for haematological cancers

1

136

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.59, 1.80]

3 Interruptions to cancer treatment (RT breaks > 2 consecutive days) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 RT to head & neck +/‐ cisplatin

1

113

Risk Ratio (M‐H, Random, 95% CI)

4.38 [0.25, 75.44]

4 Normalcy of diet (use of supplemental nutrition) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 BMT/SCT after conditioning for haematological cancers

1

136

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.55, 1.94]

Figuras y tablas -
Comparison 9. EGF versus placebo
Comparison 10. ITF versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 CT alone for colorectal cancer

1

99

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.35, 0.79]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 CT alone for colorectal cancer

1

99

Risk Ratio (M‐H, Random, 95% CI)

0.22 [0.10, 0.48]

3 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 CT alone for colorectal cancer

1

99

Risk Ratio (M‐H, Random, 95% CI)

1.52 [0.06, 36.39]

Figuras y tablas -
Comparison 10. ITF versus placebo
Comparison 11. ITF (dose comparison)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 CT alone for colorectal cancer

1

66

Risk Ratio (M‐H, Random, 95% CI)

1.3 [0.67, 2.54]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 CT alone for colorectal cancer

1

66

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.18, 3.09]

3 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 CT alone for colorectal cancer

1

66

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.90]

Figuras y tablas -
Comparison 11. ITF (dose comparison)
Comparison 12. Erythropoietin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oral mucositis (any) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 BMT/SCT after conditioning for haematological cancers

1

80

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.21, 0.60]

2 Oral mucositis (moderate + severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 BMT/SCT after conditioning for haematological cancers

1

80

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.24, 0.79]

3 Oral mucositis (severe) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 BMT/SCT after conditioning for haematological cancers

1

80

Risk Ratio (M‐H, Random, 95% CI)

0.4 [0.14, 1.17]

4 Number of days in hospital Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 BMT/SCT after conditioning for haematological cancers

1

80

Mean Difference (IV, Random, 95% CI)

‐2.95 [‐7.73, 1.83]

Figuras y tablas -
Comparison 12. Erythropoietin versus placebo