Scolaris Content Display Scolaris Content Display

Intervensi farmakologi untuk mencegah mulut kering dan disfungsi kelenjar air liur selepas radioterapi

Contraer todo Desplegar todo

Referencias

Abacioglu 1997 {unpublished data only}

Abacioglu MU. Effect of Pilocarpine for the Prevention of Radiation‐Induced Xerostomia [Dissertation]. Marmara (Turkey): Marmara University Hospital, 1997. CENTRAL

Antonadou 2002 {published data only}

Antonadou D, Pepelassi M, Synodinou M, Puglisi M, Throuvalas N. Prophylactic use of amifostine to prevent radiochemotherapy‐induced mucositis and xerostomia in head‐and‐neck cancer. International Journal of Radiation Oncology, Biology, Physics 2002;52(3):739‐47. CENTRAL

Bardet 2011 {published data only}

Bardet E, Martin L, Calais G, Alfonsi M, Feham N E, Tuchais C, et al. Subcutaneous compared with intravenous administration of amifostine in patients with head and neck cancer receiving radiotherapy: final results of the GORTEC2000‐02 phase III randomized trial. Journal of Clinical Oncology 2011;29(2):127‐33. CENTRAL
Bardet E, Martin L, Calais G, Tuchais C, Bourhis J, Rhein B, et al. Preliminary data of the GORTEC 2000‐02 phase III trial comparing intravenous and subcutaneous administration of amifostine for head and neck tumors treated by external radiotherapy. Seminars in Oncology 2002;29(6 Suppl 19):57‐60. CENTRAL

Brizel 2000 {published data only}

Brizel D, Sauer R, Wannenmacher M, Henke M, Eschwege F, Wasserman T. Randomized phase III trial of radiation amifostine in patients with head and neck cancer. 34th Annual Meeting of the American Society of Clinical Oncology; 1998 May 17‐19; Los Angeles (USA). Los Angeles (USA): American Society of Clinical Oncology, 1998. [Abs No 1487]CENTRAL
Brizel DM, Wasserman T. The influence of intravenous amifostine on xerostomia and survival during radiotherapy for head and neck cancer: two year follow‐up of a prospective randomized trial. Journal of Clinical Oncology 2004;22(14 Suppl):5536. CENTRAL
Brizel DM, Wasserman TH, Henke M, Strnad V, Rudat V, Monnier A, et al. Phase III randomized trial of amifostine as a radioprotector in head and neck cancer. Journal of Clinical Oncology 2000;18(19):3339‐45. CENTRAL
Rudat V, Meyer J, Momm F, Bendel M, Henke M, Strnad V, et al. Protective effect of amifostine on dental health after radiotherapy of the head and neck. International Journal of Radiation Oncology, Biology, Physics 2000;48(5):1339‐43. CENTRAL
Strnad V, Sauer R. Radioprotection of head and neck tissue by amifostine. Frontiers of Radiation Therapy and Oncology 2002;37:101‐11. CENTRAL
Wasserman T, Mackowiak JI, Brizel DM, Oster W, Zhang J, Peeples PJ, et al. Effect of amifostine on patient assessed clinical benefit in irradiated head and neck cancer. International Journal of Radiation Oncology, Biology, Physics 2000;48(4):1035‐9. CENTRAL
Wasserman TH, Brizel DM, Henke M, Monnier A, Eschwege F, Sauer R, et al. Influence of intravenous amifostine on xerostomia, tumor control, and survival after radiotherapy for head‐and‐ neck cancer: 2‐year follow‐up of a prospective, randomized, phase III trial. International Journal of Radiation Oncology, Biology, Physics 2005;63(4):985‐90. 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

Buentzel 2006 {published data only}

Buentzel J, Micke O, Adamietz IA, Monnier A, Glatzel M, de Vries A. Intravenous amifostine during chemoradiotherapy for head‐and‐neck cancer: a randomized placebo‐controlled phase III study. International Journal of Radiation Oncology, Biology, Physics 2006;64(3):684‐91. CENTRAL

Büntzel 1998 {published data only}

Bennett CL, Lane D, Stinson T, Glatzel M, Buntzel J. Economic analysis of amifostine as adjunctive support for patients with advanced head and neck cancer: preliminary results from a randomized phase II clinical trial from Germany. Cancer Investigation 2001;19(2):107‐13. CENTRAL
Buntzel J, Glatzel M, Schuth J, Frohlich D, Kuttner K. Selective cytoprotection with amifostine: a new strategy in supportive care of head and neck malignancies. Oto‐Rhino‐Laryngologia Nova 1997;7:276‐80. CENTRAL
Buntzel J, Kuttner K, Russell L, Oster W, Schuth J, Glatzel M. Selective cytoprotection by amifostine (A) in the treatment of head and neck cancer with simultaneous radiochemotherapy (RCT). European Cancer Conference; 1997 Sep 14‐18; Hamburg (Germany). Hamburg (Germany): ECCO, 1997. [Abs No 852]CENTRAL
Buntzel J, Kuttner K, Russell L, Oster W, Schuth J, Glatzel M. Selective cytoprotection by amifostine in the treatment of head and neck cancer with simultaneous radiochemotherapy. Proceedings of the American Society of Clinical Oncology 1997;16:393. [Abs No 1400]CENTRAL
Büntzel J. Experiences with selenium in the treatment of acute and late toxicities due to radiochemotherapy of head and neck cancer [Erfahrungen mit Natriumselenit in der Behandlung von akuten und späten Nebenwirkungen der Radiochemotherapie von Kopf‐Hals‐Karzinomen]. Medizinische Klinik 1999;94 Suppl III:49‐53. CENTRAL
Büntzel J, Glatzel M, Kuttner K, Schuth J, Oster W, Frohlich F. Selective cytoprotection with amifostine in simultaneous radiochemotherapy of head neck cancer. Annals of Oncology 1996;7(81):381. CENTRAL
Büntzel J, Glatzel M, Kuttner K, Weinaug R, Fröhlich D. Amifostine in simultaneous radiochemotherapy of advanced head and neck cancer. Seminars in Radiation Oncology 2002;12(1 Suppl 1):4‐13. CENTRAL
Büntzel J, Kuttner, Frohlich D, Schuth J, Glatzel M. Amifostine in simultaneous radiochemotherapy for head and neck cancer. Oto‐Rhino‐Laryngologia Nova 1997;7:204‐10. CENTRAL
Büntzel J, Küttner K, Fröhlich D, Glatzel M. Selective cytoprotection with amifostine in concurrent radiochemotherapy for head and neck cancer. Annals of Oncology 1998;9(5):505‐9. CENTRAL
Büntzel J, Schuth J, Küttner K, Glatzel M. Radiochemotherapy with amifostine cytoprotection for head and neck cancer. Supportive Care in Cancer 1998;6(2):155‐60. CENTRAL

Büntzel 2010 {published data only}

Buentzel J, Micke O, Glatzel M, Bruns F, Kisters K, Muecke R. Evaluation of the effect of selenium on radiation‐induced toxicities in head neck cancer patients. Journal of Clinical Oncology 2009;27(15 Suppl):e20698. CENTRAL
Büntzel J, Micke O, Glatzel M, Schäfer U, Riesenbeck D, Kisters K, et al. Selenium substitution during radiotherapy in head and neck cancer. Trace Elements and Electrolytes 2010;27:235‐9. CENTRAL
Büntzel J, Riesenbeck D, Glatzel M, Berndt‐Skorka R, Riedel T, Mücke R, et al. Limited effects of selenium substitution in the prevention of radiation‐associated toxicities. Results of a randomized study in head and neck cancer patients. Anticancer Research 2010;30(5):1829‐32. CENTRAL

Burlage 2008 {published data only}

Burlage FR, Roesink JM, Kampinga HH, Coppes RP, Terhaard C, Langendijk JA, et al. Protection of salivary function by concomitant pilocarpine during radiotherapy: a double‐blind, randomized, placebo‐controlled study. International Journal of Radiation Oncology, Biology, Physics 2008;70(1):14‐22. CENTRAL

Duncan 2005 {published data only}

Duncan GG, Epstein JB, Tu D, El Sayed S, Bezjak A, Ottaway J, et al. Quality of life, mucositis, and xerostomia from radiotherapy for head and neck cancers: a report from the NCIC CTG HN2 randomized trial of an antimicrobial lozenge to prevent mucositis. Head & Neck 2005;27(5):421‐8. CENTRAL

Fisher 2003 {published data only}

Fisher J, Scott C, Scarantino CW, Leveque FG, White RL, Rotman M, et al. Phase III quality‐of‐life study results: impact on patients' quality of life to reducing xerostomia after radiotherapy for head‐and‐neck cancer ‐ RTOG 97‐09. International Journal of Radiation Oncology, Biology, Physics 2003;56(3):832‐6. CENTRAL
Scarantino C, LeVeque F, Swann RS, White R, Schulsinger A, Hodson I, et al. Effect of pilocarpine during radiation therapy: results of RTOG 97‐09, a phase III randomized study in head and neck cancer patients. Journal of Supportive Oncology 2006;4(5):252‐8. CENTRAL
Scarantino CW, Leveque F, Scott C, White RL, Rotman M, Hodson DI, et al. A phase III study on the concurrent use of oral pilocarpine to reduce hyposalivation and mucositis associated with radiation therapy in head and neck cancer patients. Final results of RTOG 97‐09. International Journal of Radiation Oncology, Biology, Physics 2001;51(3 Suppl 1):85‐6. [Abs No 157]CENTRAL
Scarantino CW, Leveque FG, Scott CB, White RL, Rotman M, Hodson DI. A phase III study of concomitant oral pilocarpine to reduce hypo‐salivation and mucositis associated with curative radiation therapy (RT) in head and neck (H&N) cancer patients. RTOG 9709. Proceedings of the American Society of Clinical Oncology 2001;20:225a. [Abs No 897]CENTRAL

Gornitsky 2004 {published and unpublished data}

Gornitsky M, Shenouda G, Sultanem K, Katz H, Hier M, Black M, et al. Double‐blind randomized, placebo‐controlled study of pilocarpine to salvage salivary gland function during radiotherapy of patients with head and neck cancer. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontics 2004;98(1):45‐52. CENTRAL

Grötz 2001 {published data only}

Grötz KA, Henneicke‐Von Zepelin HH, Kohnen R, Kutzner J, Belz GG. Prophylaxis of mucositis and dry mouth after head and neck radiotherapy. A new treatment. Krankenhauspharmazie 2002;23(5):193‐8. CENTRAL
Grötz KA, Henneicke‐von Zepelin HH, Kohnen R, al‐Nawas B, Bockisch A, Kutzner J, et al. Prospective double‐blind study of prophylaxis of radioxerostomia with Coumarin/Troxerutine in patients with head and neck cancer [Prospektive, doppelblinde Therapiestudie zur Prophylaxe der Radioxerostomie durch Cumarin/Troxerutin bei Patienten mit Kopf‐Hals‐Karzinomen]. Strahlentherapie und Onkologie 1999;175(8):397‐404. CENTRAL
Grötz KA, Wüstenberg P, Kohnen R, Al‐Nawas B, Henneicke‐von Zepelin H, Bockisch A, et al. Prophylaxis of radiogenic sialadenitis and mucositis by coumarin/troxerutine in patients with head and neck cancer ‐ a prospective, randomized, placebo‐controlled, double‐blind study. British Journal of Oral and Maxillofacial Surgery 2001;39(1):34‐9. CENTRAL

Haddad 2002 {published data only}

Haddad P, Karimi M. A randomised, double‐blind, placebo‐controlled trial of concomitant pilocarpine with head and neck irradiation for prevention of radiation‐induced xerostomia. Radiotherapy and Oncology 2002;64(1):29‐32. CENTRAL

Haddad 2009 {published data only}

Haddad R, Sonis S, Posner M, Wirth L, Costello R, Braschayko P, et al. Randomized phase 2 study of concomitant chemoradiotherapy using weekly carboplatin/paclitaxel with or without daily subcutaneous amifostine in patients with locally advanced head and neck cancer. Cancer 2009;115(19):4514‐23. CENTRAL

Han 2010 {published data only}

Han F, Zhang JD, Shao ZY, Liu FJ. Effects of concurrent chemoradiotherapy combined with Jinlong capsules for patients with advanced nasopharyngeal squamous carcinoma. Chinese Journal of Cancer Prevention and Treatment 2010;(2):138‐9. CENTRAL

He 2004 {published data only}

He X, Hu C, Wu Y. Radioprotective effect of amifostine in nasopharyngeal carcinoma. Fudan University Journal of Medical Sciences 2004;31(1):53‐8. 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

Hu 2005 {published data only}

Hu YR, Wu CQ, Liu YJ, Wang Y, Li X, Zhong H, et al. Clinical observation on effect of shenqi fanghou recipe in preventing and treating radiation injury in patients with head and neck tumour. Zhongguo Zhong Xi Yi Jie He Za Zhi 2005;25(7):623‐5. CENTRAL

Jaguar 2015 {published data only}

Jaguar GC. A Prospective Study of Bethanechol in the Salivary Glands Physiology in Patients Following Head and Neck Radiotherapy [Estudo Prospectivo do Uso do Betanecol na Fisiologia de Glândulas Salivares em Pacientes Irradiados em Região de Cabeça e Pescoço] [Dissertation]. Sao Paulo (Brazil): Fundaçao Antonio Prudente, 2010. CENTRAL
Jaguar GC, Campos L, Pellizzon AC, Lima EN, Kowalski LP, Alves FA. Double‐blind randomized prospective trial of bethanechol in the prevention of radiation‐induced salivary gland dysfunction in cancer patients. Supportive Care in Cancer 2012;20(1 Suppl):S252. CENTRAL
Jaguar GC, Lima EN, Kowalski LP, Pellizzon AC, Carvalho AL, Boccaletti KW, et al. Double blind randomized prospective trial of bethanechol in the prevention of radiation‐induced salivary gland dysfunction in head and neck cancer patients. Radiotherapy and Oncology 2015;115(2):253‐6. CENTRAL

Jellema 2006 {published data only}

Jellema AP, Slotman BJ, Muller MJ, Leemans CR, Smeele LE, Hoekman K, et al. Radiotherapy alone, versus radiotherapy with amifostine 3 times weekly, versus radiotherapy with amifostine 5 times weekly: a prospective randomized study in squamous cell head and neck cancer. Cancer 2006;107(3):544‐53. CENTRAL

Jham 2007 {published data only}

Jham BC, Chen H, Carvalho AL, Freire AR. A randomized phase III prospective trial of bethanechol to prevent mucositis, candidiasis, and taste loss in patients with head and neck cancer undergoing radiotherapy: a secondary analysis. Journal of Oral Science 2009;51(4):565‐72. CENTRAL
Jham BC, Teixeira IV, Aboud CG, Carvalho AL, Coelho Mde M, Freire AR. A randomized phase III prospective trial of bethanechol to prevent radiotherapy‐induced salivary gland damage in patients with head and neck cancer. Oral Oncology 2007;43(2):137‐42. CENTRAL

Lajtman 2000 {published data only}

Lajtman Z, Krajina Z, Krpan D, Vincelj J, Borcic V, Popovic‐Kovacic J. Pilocarpine in the prevention of postirradiation xerostomia. Acta Medica Croatica 2000;54(2):65‐7. CENTRAL

Lanzós 2010 {published data only}

Lanzós I, Herrera D, Santos S, O'Connor A, Peña C, Lanzós E, et al. Mucositis in irradiated cancer patients: effects of an antiseptic mouthrinse. Medicina Oral, Patología Oral y Cirugía Bucal 2010;15(5):e732‐8. CENTRAL

Le 2011 {published data only}

Le QT, Kim HE, Schneider CJ, Murakozy 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

Lin 2014 {published data only}

Lin KS, Jen YM, Chao TY, Lin YS, Wang LH, Lin CJ, et al. Prevention of acute radiation‐associated toxicity by traditional chinese medicine Tianwang Buxin mini‐pills in patients with head and neck cancer. Journal of Medical Sciences (Taipei, Taiwan) 2014;34:152‐60. CENTRAL

Lozada‐Nur 1998 {published and unpublished data}

Lozada‐Nur F, Schoelch M, Fu K, Muscoplat C, Trivedi M, Smith C, et al. A pilot study to evaluate the effect of pilocarpine tablets on salivary flow and mucositis in head and neck cancer patients during radiotherapy. Proceedings of the American Society of Clinical Oncology 1998;17:399. [Abs No 1541]CENTRAL

Patni 2004 {published and unpublished data}

Patni N, Patni S, Bapna A, Somani N, Gupta A, Ratnam BV. The role of amifostine in prophylaxis of radiotherapy induced mucositis and xerostomia in head and neck cancer. Journal of Clinical Oncology 2004;22(14 Suppl):5568. CENTRAL

Peng 2006 {published data only}

Peng LH, Sun Y, Zhang XL, Zhang Q, Fu S. Efficacy of amifostine on patients with head and neck squamous cell carcinomas undergoing radiation therapy and chemotherapy and clinical observation on its hematologic toxicity. Journal of China Pharmacy 2006;17(5):1166‐8. CENTRAL

Pimentel 2014 {published data only}

Pimentel MJ, Filho MM, Araújo M, Gomes DQ, Da Costa LJ. Evaluation of radioprotective effect of pilocarpine ingestion on salivary glands. Anticancer Research 2014;34(4):1993‐9. CENTRAL

Reshma 2012 {published data only}

Reshma K, Kamalaksh S, Bindu YS, Pramod K, Asfa A, Amritha D, et al. Tulasi (Ocimum Sanctum) as radioprotector in head and neck cancer patients undergoing radiation therapy. Biomedicine 2012;32:39‐44. CENTRAL

Rode 1999 {published data only}

Rode M, Smid L, Budihna M, Soba E, Rode M, Gaspersic D. The effect of pilocarpine and biperiden on salivary secretion during and after radiotherapy in head and neck cancer patients. International Journal of Radiation Oncology, Biology, Physics 1999;45(2):373‐8. CENTRAL

Sangthawan 2001 {published data only}

Sangthawan D, Watthanaarpornchai S, Phungrassami T. Randomized double blind, placebo‐controlled study of pilocarpine administered during head and neck irradiation to reduce xerostomia. Journal of the Medical Association of Thailand 2001;84(2):195‐203. CENTRAL
Sangthawan D, Watthanaarpornchai S, Phungrassami T. Randomized double blind, placebo‐controlled study of pilocarpine administered during head and neck irradiation to reduce xerostomia. Radiation Oncology Association of Thailand 2002;7:242. CENTRAL

Vacha 2003 {published data only}

Vacha P, Fehlauer F, Mahlmann B, Marx M, Hinke A, Sommer K, et al. Randomized phase III trial of postoperative radiochemotherapy +/‐ amifostine in head and neck cancer. Is there evidence for radioprotection?. Strahlentherapie und Onkologie 2003;179(6):385‐9. CENTRAL
Vacha P, Marx M, Engel A, Richter E, Feyerabend T. Side effects of postoperative radiochemotherapy with amifostine versus radiochemotherapy alone in head and neck tumors. Preliminary results of a prospective randomized trial. Strahlentherapie und Onkologie 1999;175 Suppl 4:18‐22. CENTRAL

Valdez 1993 {published data only}

Valdez IH, Wolff A, Atkinson JC, Macynski AA, Fox PC. Use of pilocarpine during head and neck radiation therapy to reduce xerostomia and salivary dysfunction. Cancer 1993;71(5):1848‐51. CENTRAL
Wolff A, Atkinson JC, Macynski AA, Fox PC. Oral complications of cancer therapies. Pretherapy interventions to modify salivary dysfunction. NCI Monographs 1990;(9):87‐90. CENTRAL

Veerasarn 2006 {published and unpublished data}

Veerasarn V, Phromratanapongse P, Suntornpong N, Lorvidhaya V, Chitapanarux I, Tesavibul C, et al. Effect of amifostine to prevent radiotherapy‐induced acute and late toxicity in head and neck cancer patients who had normal or mild impaired salivary gland dysfunction. European Journal of Cancer Supplements 2003;1(5):S273. [Abs No 908]CENTRAL
Veerasarn V, Phromratanapongse P, Suntornpong N, Lorvidhaya V, Sukthomya V, Chitapanarux I, et al. Effect of amifostine to prevent radiotherapy‐induced acute and late toxicity in head and neck cancer patients who had normal or mild impaired salivary gland function. Journal of the Medical Association of Thailand 2006;89(12):2056‐67. CENTRAL

Wang 1998 {published data only}

Wang Q, Liu H, Qiao N. Effects of traditional Chinese medicine on salivary glands in the patients with head and neck cancer during radiotherapy. Zhongguo Zhong Xi Yi Jie He Za Zhi 1998;18(11):662‐4. CENTRAL

Warde 2002 {published data only}

Warde P, O'Sullivan B, Aslanidis J, Kroll B, Lockwood G, Math M, et al. A phase III placebo‐controlled trial of oral pilocarpine in patients undergoing radiotherapy for head and neck cancer. International Journal of Radiation Oncology, Biology, Physics 2002;54(1):9‐13. CENTRAL

Watanabe 2010 {published data only}

Watanabe T, Ishihara M, Matsuura K, Mizuta K, Itoh Y. Polaprezinc prevents oral mucositis associated with radiochemotherapy in patients with head and neck cancer. International Journal of Cancer 2010;127(8):1984‐90. CENTRAL

Anné 2002 {published data only}

Anné PR. Phase II trial of subcutaneous amifostine in patients undergoing radiation therapy for head and neck cancer. Seminars in Oncology 2002;29(6 Suppl 19):80‐3. CENTRAL

Bagga 2007 {published data only}

Bagga P, Anand AK, Raina A, Choudhary PS, Chaudhoory AR. Evaluation of pilocarpine for the prevention of radiation induced xerostomia in head and neck cancers. Annals of Oncology 2007;18(Suppl 9):ix177. [Abs No 62]CENTRAL

Bakowski 1978 {published data only}

Bakowski MT, Macdonald E, Mould RF, Cawte P, Sloggem J, Barrett A, et al. Double blind controlled clinical trial of radiation plus razoxane (ICRF 159) versus radiation plus placebo in the treatment of head and neck cancer. International Journal of Radiation Oncology, Biology, Physics 1978;4(1‐2):115‐9. CENTRAL

Belcaro 2008 {published data only}

Belcaro G, Cesarone MR, Genovesi D, Ledda A, Vinciguerra G, Ricci A, et al. Pycnogenol may alleviate adverse effects in oncologic treatment. Panminerva Medica 2008;50(3):227‐34. CENTRAL

Bohuslavizki 1998 {published data only}

Bohuslavizki KH, Klutmann S, Bleckmann C, Brenner W, Lassmann S, Mester J, et al. Salivary gland protection by amifostine in high‐dose radioiodine therapy of differentiated thyroid cancer. Strahlentherapie und Onkologie 1999;175(2):57‐61. CENTRAL
Bohuslavizki KH, Klutmann S, Brenner W, Kröger S, Buchert R, Bleckmann C, et al. Radioprotection of salivary glands by amifostine in high‐dose radioiodine treatment. Results of a double‐blinded, placebo‐controlled study in patients with differentiated thyroid cancer. Strahlentherapie und Onkologie 1999;175(Suppl 4):6‐12. CENTRAL
Bohuslavizki KH, Klutmann S, Brenner W, Mester J, Henze E, Clausen M. Salivary gland protection by amifostine in high‐dose radioiodine treatment: results of a double‐blind placebo‐controlled study. Journal of Clinical Oncology 1998;16(11):3542‐9. CENTRAL
Bohuslavizki KH, Klutmann S, Jenicke L, Kröger S, Buchert R, Mester J, et al. Salivary gland protection by S‐2‐(3‐amino‐propylamino)‐ethylphosphorothioic acid (amifostine) in high‐dose radioiodine treatment: results obtained in a rabbit animal model and in a double‐blind multi‐arm trial. Cancer Biotherapy & Radiopharmaceuticals 1999;14(5):337‐47. CENTRAL

Borg 2007 {published data only}

Borg M, Krishnan S, Olver L, Stein B, Chatterton B, Coates E, et al. Randomised double‐blind trial of amifostine vs placebo for radiation‐induced xerostomia in patients with head and neck cancer. ANZ Journal of Surgery 2007;77(12):A3. CENTRAL

Bourhis 2000 {published data only}

Bourhis J, de Crevoisier R, Abdulkarim B, Deutsch E, Lusinchi A, Luboinski B, et al. A randomized study of very accelerated radiotherapy with and without amifostine in head and neck squamous cell carcinoma. International Journal of Radiation Oncology, Biology, Physics 2000;46(5):1105‐8. CENTRAL

Braaksma 2002 {published data only}

Braaksma M, Levendag P. Tools for optimal tissue sparing in concomitant chemoradiation of advanced head and neck cancer: subcutaneous amifostine and computed tomography‐based target delineation. Seminars in Oncology 2002;29(6 Suppl 19):63‐70. CENTRAL

Braaksma 2005 {published data only (unpublished sought but not used)}

Braaksma M, van Agthoven M, Nijdam W, Uyl‐de Groot C, Levendag P. Costs of treatment intensification for head and neck cancer: concomitant chemoradiation randomised for radioprotection with amifostine. European Journal of Cancer 2005;41(14):2102‐11. CENTRAL

Chambers 2005 {published data only}

Chambers MS, Posner MR, Jones CU, Weber RS, Vitti R. Two phase III clinical studies of cevimeline for post‐radiation xerostomia in patients with head and neck cancer. Journal of Clinical Oncology 2005;23(16 Suppl):5503. CENTRAL

Demiroz 2012 {published data only}

Demiroz C, Ozcan L, Cebelli G, Karadag O, Ozsahin EM. The effect of amifostine on acute and late radiation side effects in head and neck cancer patients. Turkiye Klinikleri Journal of Medical Sciences 2012;32:1207‐16. CENTRAL

Fallahi 2013 {published data only}

Fallahi B, Beiki D, Abedi SM, Saghari M, Fard‐Esfahani A, Akhzari F, et al. Does vitamin E protect salivary glands from I‐131 radiation damage in patients with thyroid cancer?. Nuclear Medicine Communications 2013;34(8):777‐86. CENTRAL

Fan 2011 {published data only}

Fan Z, Lin H, Liu Z, He Q, Zhang S, Li W. Protective effect of amifostine in irradiation mucositis of the oral cavity and dry mouth after nasapharyngeal carcinoma. Military Medical Journal of Southeast China 2011;13(2):146‐8. CENTRAL

Franzén 1995 {published data only}

Franzén L, Henriksson R, Littbrand B, Zackrisson B. Effects of sucralfate on mucositis during and following radiotherapy of malignancies in the head and neck region. A double‐blind placebo‐controlled study. Acta Oncologica (Stockholm, Sweden) 1995;34(2):219‐23. CENTRAL

Fuertes 2004 {published data only}

Fuertes Cabero S, Setoain Perego X, Rovirosa Casino A, Mateos Fernández JJ, Fuster Pelfort D, Ferre Jorge J, et al. Usefulness of pilocarpine in the prevention of xerostomia in patients with head and neck cancer treated with radiotherapy. Assessment with gammagraphy and salivary flow [Utilidad de la pilocarpina como profiláctico de xerostomia en pacientes con cáncer de cabeza y cuello tratados con radioterapia. Valoración mediante gammagrafía y flujo salivar]. Revista Española de Medicina Nuclear 2004;23(4):259‐66. CENTRAL

Goyal 2007 {published data only}

Goyal S, Sharma DN, Julka PK, Rath GK. Effect of oral pilocarpine on xerostomia and quality of life in patients receiving curative radiotherapy for head and neck cancers. Journal of Clinical Oncology 2007;25(18 Suppl):6088. CENTRAL

Gu 2014 {published data only}

Gu J, Zhu S, Li X, Wu H, Li Y, Hua F. Effect of amifostine in head and neck cancer patients treated with radiotherapy: a systematic review and meta‐analysis based on randomized controlled trials. PloS One 2014;9(5):e95968. CENTRAL

Johnson 2002 {published data only}

Johnson DJ, Scott CB, Marks JE, Seay TE, Atkins JN, Berk LB, et al. Assessment of quality of life and oral function of patients participating in a phase II study of radioprotection of oral and pharyngeal mucosa by the prostaglandin E(1) analog misoprostol (RTOG 96‐07). International Journal of Radiation Oncology, Biology, Physics 2002;54(5):1455‐9. CENTRAL

Karacetin 2004 {published data only}

Karacetin D, Yücel B, Leblebicioglu B, Aksakal O, Maral O, Incekara O. A randomized trial of amifostine as radioprotector in the radiotherapy of head and neck cancer. Journal of BUON 2004;9(1):23‐6. CENTRAL

Koukourakis 2000 {published data only}

Koukourakis MI, Kyrias G, Kakolyris S, Kouroussis C, Frangiadaki C, Giatromanolaki A, et al. Subcutaneous administration of amifostine during fractionated radiotherapy: a randomized phase II study. Journal of Clinical Oncology 2000;18(11):2226‐33. CENTRAL

Kumarchandra 2010 {published data only}

Kumarchandra R, Shenoy K, Bindu YS, Kadaba P, Chandrashekar R. Ocimum Sanctum as a radioprotector in head and neck cancer patients undergoing radiation therapy. Free Radical Biology and Medicine 2010;49 Suppl:S64. CENTRAL

Manoor 2014 {published data only}

Manoor Maiya V, Vaid N, Basu S, Vatyam S, Hegde S, Deshmukh S, et al. The use of xylitol for the prevention of xerostomia in patients receiving intensity modulated radiation therapy for head and neck cancers. International Journal of Radiation Oncology, Biology, Physics 2014;90(1 Suppl):S562. CENTRAL

Mateos 2001 {published data only}

Mateos JJ, Setoain X, Ferre J, Rovirosa A, Navalpotro B, Martin F, et al. Salivary scintigraphy for assessing the protective effect of pilocarpine in head and neck irradiated tumours. Nuclear Medicine Communications 2001;22(6):651‐6. CENTRAL

Mitine 2000 {published data only}

Mitine C, Chaltin M, Salembier C, Merlo P, Beauduin M. Head and neck radiotherapy: does pilocarpine hydrochloride reduce radiation‐induced xerostomia?. Proceedings of the management of head and neck tumors: what are the challenges for the third millennium; 1999 Dec 3‐4; Brussels (Belgium). European Laryngological Society, 1999. CENTRAL

Mix 2013 {published data only}

Mix MD, Jameson M, Tills M, Dibaj S, Groman A, Jaggernauth W, et al. Randomized double‐blind, placebo‐controlled, multicenter phase 2 trial of selenomethionine as a modulator of efficacy and toxicity of chemoradiation in locally‐advanced squamous cell carcinoma of the head and neck. International Journal of Radiation Oncology, Biology, Physics 2013;87(2 Suppl):S466‐7. CENTRAL

Nicolatou‐Galitis 2003 {published data only}

Nicolatou‐Galitis O, Sotiropoulou‐Lontou A, Velegraki A, Pissakas G, Kolitsi G, Kyprianou K, et al. Oral candidiasis in head and neck cancer patients receiving radiotherapy with amifostine cytoprotection. Oral Oncology 2003;39:397‐401. CENTRAL

Norberg‐Spaak 1996 {published data only}

Norberg‐Spaak LE, Lundquist PG, Berndtson M, Klintenberg C. Can pilocarpine treatment during irradiation prevent salivary gland damage?. Swedish Dental Journal 1996;20:234. CENTRAL

Norberg‐Spaak 1997 {published data only}

Norberg‐Spaak LE. Can pilocarpine treatment during irradiation prevent salivary gland damage. Clinical Otolaryngology and Allied Sciences 1997;22:88. CENTRAL

Nyárády 2006 {unpublished data only}

Nyárády Z, Németh A, Bán A, Mukics A, Nyárády J, Ember I, et al. A randomized study to assess the effectiveness of orally administered pilocarpine during and after radiotherapy of head and neck cancer. Anticancer Research 2006;26(2B):1557‐62. CENTRAL
Nyárády Z, Németh Á, Mukics A, Olasz L. Relieving symptoms of xerostomia with oral pilocarpine (Salagen) during irradiation in head‐and‐neck cancer. Journal of Cranio‐Maxillo‐Facial Surgery 2004;32 Suppl 1:230. [Abs No 459]CENTRAL

Park 2012 {published data only}

Park J, McGuire DB, Kang H. Effects of cold sterile normal saline (CSNS) mouth care in head and neck cancer (HNC) patients undergoing concurrent chemoradiotherapy (CCRT). Supportive Care in Cancer 2012;20(1 Suppl):S246‐7. [Abs No 1028]CENTRAL

Park 2012a {published data only}

Park K‐N, Son Y‐I, Baek CH. Protection of radiotherapy‐induced xerostomia with vitamin E+C complex. Otolaryngology ‐ Head and Neck Surgery 2012;147(2 Suppl):P174. CENTRAL

Peters 1999 {published data only}

Peters K, Mücke R, Hamann D, Ziegler PG, Fietkau R. Supportive use of amifostine in patients with head and neck tumours undergoing radio‐chemotherapy. Is it possible to limit the duration of the application of amifostine?. Strahlentherapie und Onkologie 1999;175 Suppl 4:23‐6. CENTRAL

Qian 2003 {published data only}

Qian Y. Effects of Traditional Chinese Medicine on Salivary Glands of Patients with Head and Neck Cancer During Radiotherapy [Dissertation]. China National Knowledge Infrastructure (CNKI), 2003. CENTRAL

Resubal 2011 {published data only}

Resubal JR, Calaguas MJ. The effect of zilongjin(r) in patients with head and neck cancer needing radiotherapy with and without chemotherapy. Radiotherapy and Oncology 2011;99(Suppl 1):S336. CENTRAL

Rieger 2012 {published data only}

Rieger JM, Jha N, Lam Tang JA, Harris J, Seikaly H. Functional outcomes related to the prevention of radiation‐induced xerostomia: oral pilocarpine versus submandibular salivary gland transfer. Head & Neck 2012;34(2):168‐74. CENTRAL

Rischin 2010 {published data only}

Rischin D, Peters LJ, O'Sullivan B, Giralt J, Fisher R, Yuen K, et al. Tirapazamine, cisplatin, and radiation versus cisplatin and radiation for advanced squamous cell carcinoma of the head and neck (TROG 02.02, HeadSTART): a phase III trial of the Trans‐Tasman Radiation Oncology Group. Journal of Clinical Oncology 2010;28(18):2989‐95. CENTRAL

Rudat 2005 {published data only}

Rudat V, Münter M, Rades D, Grötz K, Haberkorn U, Brenner W. The effect of amifostine or IMRT to preserve the parotid function after radiotherapy of the head and neck region measured by quantitative salivary gland scintigraphy. Journal of Clinical Oncology 2005;23(16 Suppl):5502. CENTRAL

Schönekäs 1999 {published data only}

Schönekäs KG, Wagner W, Prott FJ. Amifostine ‐ a radioprotector in locally advanced head and neck tumours. Strahlentherapie und Onkologie 1999;175 Suppl 4:27‐9. CENTRAL

Sharma 2012 {published data only}

Sharma A, Rath GK, Chaudhary SP, Thakar A, Mohanti BK, Bahadur S. Lactobacillus brevis CD2 lozenges reduce radiation‐ and chemotherapy‐induced mucositis in patients with head and neck cancer: a randomized double‐blind placebo‐controlled study. European Journal of Cancer 2012;48(6):875‐81. CENTRAL

Strnad 1997 {published data only}

Strnad V, Sauer R, Krafft T, Lerch S. Preliminary results of a randomised study using WR‐2721 in radiation therapy alone in patients with head and neck cancer. European Journal of Cancer 1997;33(Suppl 8):S189. CENTRAL

Su 2006 {published data only}

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 (Sudbury, Mass.) 2006;12(3):182‐8. CENTRAL

Takahashi 1986 {published data only}

Takahashi I, Nagai T, Miyaishi K, Maehara Y, Niibe H. Clinical study of the radioprotective effects of amifostine (YM‐08310, WR‐2721) on chronic radiation injury. International Journal of Radiation Oncology, Biology, Physics 1986;12(6):935‐8. CENTRAL

Thorstad 2003 {published data only}

Thorstad WL, Haughey B, Chao KS. Pilot study of subcutaneous amifostine in patients undergoing postoperative intensity modulated radiation therapy for head and neck cancer: preliminary data. Seminars in Oncology 2003;30(6 Suppl 18):96‐100. CENTRAL

Uchiyama 2005 {published data only}

Uchiyama Y, Murakami S, Kakimoto N, Nakatani A, Furukawa S. Effectiveness of cepharanthin in decreasing interruptions during radiation therapy for oral cancer. Oral Radiology 2005;21(1):41‐4. CENTRAL

Zale 1993 {published data only}

Zale M, Chambers MS, Khan Z. Pilocarpine effects on xerostomia associated with radiation therapy. Journal of Dental Research 1993;72(Abs Spec Issue):375. [Abs No 2175]CENTRAL

Zimmerman 1997 {published data only}

Zimmerman RP, Mark RJ, Juillard GF. Timing of pilocarpine treatment during head and neck radiotherapy: concomitant administration reduces xerostomia better than post‐radiation pilocarpine. International Journal of Radiation Oncology, Biology, Physics 1996;36 Suppl 1(1):236. [Abs No 155]CENTRAL
Zimmerman RP, Mark RJ, Tran LM, Juillard GF. Concomitant pilocarpine during head and neck irradiation is associated with decreased posttreatment xerostomia. International Journal of Radiation Oncology, Biology, Physics 1997;37(3):571‐5. CENTRAL
Zimmerman RP, Rufus JM, Juillard GF. Concomitant pilocarpine during head and neck irradiation reduces xerostomia. Supportive Care in Cancer 1996;4(3):243. [Abs No 76]CENTRAL

Yu 2009 {published data only}

Yu DS, Zhao W, Wu XL, Huang ZY, Huang RB. Amifostine in prevention of radiation‐induced xerostomia. Journal of Sun Yat‐Sen University (Medical Sciences) 2009;30(4S):86‐7, 95. CENTRAL

NCT02430298 {published data only}

NCT02430298. Topical/oral melatonin for preventing concurrent radiochemotherapy induced oral mucositis/xerostomia cancer patients [Topical and oral melatonin for preventing concurrent radiochemotherapy induced oral mucositis and xerostomia in head and neck cancer patients]. clinicaltrials.gov/show/NCT02430298 (first received 28 January 2014). CENTRAL

Ahlner 1994

Ahlner BH, Hagelqvist E, Lind MG. Influence on rabbit submandibular gland injury by stimulation or inhibition of gland function during irradiation. Histology and morphometry after 15 gray. Annals of Otology, Rhinology, and Laryngology 1994;103(2):125‐34.

Andreassen 2003

Andreassen CN, Grau C, Lindegaard JC. Chemical radioprotection: a critical review of amifostine as a cytoprotector in radiotherapy. Seminars in Radiation Oncology 2003;13(1):62‐72.

Bennett 2001

Bennett CL, Lane D, Stinson T, Glatzel M, Buntzel J. Economic analysis of amifostine as adjunctive support for patients with advanced head and neck cancer: preliminary results from a randomized phase II clinical trial from Germany. Cancer Investigations 2001;19(2):107‐13.

Bohuslavizki 1998

Bohuslavizki KH, Brenner W, Klutmann S, Hübner RH, Lassmann S, Feyerabend B, et al. Radioprotection of salivary glands by amifostine in high‐dose radioiodine therapy. Journal of Nuclear Medicine 1998;39(7):1237‐42.

Buglione 2016

Buglione M, Cavagnini R, Di Rosario F, Maddalo M, Vassalli L, Grisanti S, et al. Oral toxicity management in head and neck cancer patients treated with chemotherapy and radiation: Xerostomia and trismus (Part 2). Literature review and consensus statement. Critical Reviews in Oncology/Hematology 2016;102:47‐54.

Clarkson 2010

Clarkson JE, Worthington HV, Furness S, McCabe M, Khalid T, Meyer S. Interventions for treating oral mucositis for patients with cancer receiving treatment. Cochrane Database of Systematic Reviews 2010, Issue 8. [DOI: 10.1002/14651858.CD001973.pub4]

Davies 2015

Davies AN, Thompson J. Parasympathomimetic drugs for the treatment of salivary gland dysfunction due to radiotherapy. Cochrane Database of Systematic Reviews 2015, Issue 10. [DOI: 10.1002/14651858.CD003782.pub3]

Dawes 1987

Dawes C. Physiological factors affecting salivary flow rate, oral sugar clearance, and the sensation of dry mouth in man. Journal of Dental Research 1987;66 Spec No:648‐53.

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.

Epstein 1994

Epstein JB, Burchell JL, Emerton S, Le ND, Silverman S. A clinical trial of bethanechol in patients with xerostomia after radiation therapy. A pilot study. Oral Surgery, Oral Medicine, and Oral Pathology 1994;77(6):610‐4.

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.

Guchelaar 1997

Guchelaar HJ, Vermes A, Meerwaldt JH. Radiation‐induced xerostomia: pathophysiology, clinical course and supportive treatment. Supportive Care in Cancer 1997;5(4):281‐8.

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.

Kontopantelis 2012

Kontopantelis E, Reeves D. Performance of statistical methods for meta‐analysis when true study effects are non‐normally distributed: A simulation study. Statistical Methods in Medical Research 2012;21(4):409‐26.

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.

Mercadante 2017

Mercadante V, Al Hamad A, Lodi G, Porter S, Fedele S. Interventions for the management of radiotherapy‐induced xerostomia and hyposalivation: A systematic review and meta‐analysis. Oral Oncology 2017;66:64‐74.

Navazesh 1992

Navazesh M, Christensen C, Brightman V. Clinical criteria for the diagnosis of salivary gland hypofunction. Journal of Dental Research 1992;71(7):1363‐9.

Pankhurst 1996

Pankhurst CL, Smith EC, Rogers JO, Dunne SM, Jackson SH, Proctor G. Diagnosis and management of the dry mouth: Part 1. Dental Update 1996;23(2):56‐62.

Porter 2004

Porter SR, Scully C, Hegarty AM. An update of the etiology and management of xerostomia. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontics 2004;97(1):28‐46.

Rades 2004

Rades D, Fehlauer F, Bajrovic A, Mahlmann B, Richter E, Alberti W. Serious adverse effects of amifostine during radiotherapy in head and neck cancer patients. Radiotherapy and Oncology 2004;70(3):261‐4.

Rode 2001

Rode M, Smid L, Budihna M, Gassperssic D, Rode M, Soba E. The influence of pilocarpine and biperiden on pH value and calcium, phosphate, and bicarbonate concentrations in saliva during and after radiotherapy for head and neck cancer. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontics 2001;92(5):509‐14.

Shiboski 2007

Shiboski CH, Hodgson TA, Ship JA, Schiodt M. Management of salivary hypofunction during and after radiotherapy. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontics 2007;103 Suppl(S66):e1‐19.

Sreebny 1996

Sreebny LM. Xerostomia: diagnosis, management and clinical complications. In: Edgar WM, O'Mullane DM editor(s). Saliva and Oral Health. 2nd Edition. London: British Dental Association, 1996:43‐66.

Takahashi 1986

Takahashi T, Nagai K, Miyaishi K, Maehara Y, Niibe H. Clinical study of the radioprotective effects of amifostine (TM‐08310, WR‐2721) on chronic radiation injury. International Journal of Radiation Oncology, Biology, Physics 1986;12:935‐8.

Wiseman 1995

Wiseman LR, Faulds D. Oral pilocarpine: a review of its pharmacological properties and clinical potential in xerostomia. Drugs 1995;49(1):143‐55.

Worthington 2011

Worthington HV, Clarkson JE, Bryan G, Furness S, Glenny A‐M, 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]

Zimmerman 1997

Zimmerman RP, Mark RJ, Tran LM, Juillard GF. Concomitant pilocarpine during head and neck irradiation is associated with decreased posttreatment xerostomia. International Journal of Radiation Oncology, Biology, Physics 1997;37(3):571‐5.

Tavender 2004

Tavender E, Davies AN, Glenny AM. Pharmacological interventions for preventing salivary gland dysfunction following radiotherapy. Cochrane Database of Systematic Reviews 2004, Issue 3. [DOI: 10.1002/14651858.CD004940]

Tavender 2012

Tavender E, Davies AN, Glenny AM. Pharmacological interventions for preventing salivary gland dysfunction following radiotherapy. Cochrane Database of Systematic Reviews 2012, Issue 6. [DOI: 10.1002/14651858.CD004940.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Abacioglu 1997

Methods

Location: Turkey

Number of centres: 1

Date of enrolment: July 1996 to January 1997

Participants

Inclusion criteria: aged between 18 to 70 years. Histopathologic diagnosis of SCC of head and neck (nasopharynx, larynx, oropharynx, hypopharynx, oral cavity). WHO performance status 0 to 2. Patients to receive primary or postoperative radiation treatment for a minimum of 46 Gy totally and treatment fields to include at least the tail of parotis (1/3), submandibular glands and part of sublingual and minor salivary glands

Exclusion criteria: patients with a histopathologic diagnosis other than SCC. Patients with an autoimmune disorder (e.g. Sjögren Syndrome) or diseases effecting saliva secretion. Difficulty in co‐operation for saliva collection, understanding the questionnaire and attending the follow‐up visits

Age (years): pilocarpine: median 55 years, range 38 to 68 years; control: median 50 years, range 30 to 61 years

Gender (M:F): pilocarpine 12:0; control 11:1

Cancer type: tumour location: pilocarpine: larynx = 8, nasopharynx = 2 and oral cavity = 2; control: larynx = 7, nasopharynx = 4 and oral cavity = 1

Radiotherapy: pilocarpine: mean dose = 60.2 Gy (range 48 to 70 Gy), number of fractions = 30.1 (mean), treatment time = 44.9 days (mean); control: mean dose = 63.8 Gy (range 50 to 70 Gy), number of fractions = 31.9 (mean), treatment time = 48.2 days (mean)

Chemotherapy: none

Number randomised: 24 (12 per group)

Number evaluated: 24 (no dropouts, although not all participants available at all time points)

Interventions

Pilocarpine versus no intervention

Pilocarpine: 5 mg 3 times daily (4% solution) for 3 months from the beginning of RT

Control: no treatment

Outcomes

Xerostomia: subjective evaluation scores for xerostomia (0 = no symptoms, 11 = severe xerostomia). Questionnaire included 5 questions

Salivary flow rates: unstimulated and stimulated whole saliva secretion (unstimulated saliva pH measurements also recorded)

Adverse effects: no serious toxicity

Survival data: not reported

Other oral symptoms: not reported

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: before RT, during RT, end of RT and 3 months after start of RT

Funding

None

Trial registration

Not registered nor published

Sample size calculation presented

Not included

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Not explicit in trial report. Comment from author: "randomisation was performed with block randomisation with stratification of treatment fields"

Allocation concealment (selection bias)

Low risk

Comment from author: "sealed envelope were used for concealing"

Blinding (performance bias and detection bias)
patients/carers

High risk

Pilocarpine versus no intervention. Blinding not possible

Blinding (performance bias and detection bias)
outcome assessment

High risk

Not possible due to 'no intervention' group and subjective assessment of xerostomia

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment from author: "no dropouts"

Number of participants available for assessment varies by time point, however, those missing for assessment unlikely to influence results

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Antonadou 2002

Methods

Location: Greece

Number of centres: 1

Date of enrolment: January 1997 to January 1998

Participants

Inclusion criteria: histologically proven squamous cell carcinoma of the head and neck. A primary tumour greater than or = T2N0M0, expected survival time greater than or = 12 months, no evidence of metastasis, and no prior chemotherapy or RT. Normal liver and kidney function, adequate bone marrow reserve, no current or previous history of cardiovascular disease and no active systemic infection

Exclusion criteria: not reported

Age (years): amifostine: mean 53.3 (SD 6.9); control: mean 60.3 (SD 5.5)

Gender: amifostine: 13 M, 9 F; control: 16 M, 7 F

Cancer type: tumour location: (amifostine/control) nasopharynx = 2/3, oral cavity = 9/11, larynx = 6/6 and oropharynx = 5/3. TNM classification: (amifostine/control) T2 = 6/6, T3 = 13/16, T4 = 3/1, N0 = 12/14 and N1 to 3 = 10/9

Radiotherapy: mean total dose = amifostine: 66.8 Gy (SD 3.2); control: 66.4 Gy (SD 3.4). Treatment duration: mean = amifostine: 49.6 (SD 4.5) days; control: 55.9 (SD 8.9)

Chemotherapy: carboplatin (90 mg/m²), once a week before RT in both groups

Number randomised: 50 (amifostine 25, control 25)

Number evaluated: 45 (amifostine 22, control 23)

Interventions

Amifostine versus no intervention

Amifostine (300 mg/m2), IV 30 minutes before RT on days 1 to 5 of each week. Antimetic treatment administered IV before the amifostine
Control: nothing

Outcomes

Xerostomia: incidence of late xerostomia (RTOG grade 2 or more ‐ measured on a 0 to 4 scale)

Salivary flow rates: not reported

Adverse effects: haematologic toxicity, nausea, vomiting and transient hypotension

Survival data: progression‐free survival at 18 months

Other oral symptoms: incidence of grade 3 or greater acute mucositis and dysphagia

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: xerostomia: 3, 6, 9, 12 and 18 months after RT; haematologic toxicity and acute non‐haematological toxicities (mucositis and dysphagia): weekly for 7 weeks during RT, then 1, 2 and 3 months after RT

Funding

Not reported

Trial registration

Not registered

Sample size calculation presented

Yes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomized (1:1)"

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding (performance bias and detection bias)
patients/carers

High risk

Amifostine versus no intervention. Blinding not possible

Blinding (performance bias and detection bias)
outcome assessment

High risk

Not possible due to 'no intervention' group and subjective assessment of xerostomia

Incomplete outcome data (attrition bias)
All outcomes

Low risk

45/50 participants evaluated (equal dropouts between groups). 3 participants dropped out of the amifostine arm. 2 denied further treatment (1 = week 2 and 1 = week 4) and 1 was lost to follow‐up. In the control arm, 1 participant died and 1 received palliative treatment because of disease progression

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Bardet 2011

Methods

Location: France

Number of centres: 27

Date of enrolment: March 2001 to January 2006

Participants

Inclusion criteria: newly diagnosed head and neck, eligible for radiotherapy. Over 75% of both parotid glands in field. Performance status ≤ 2, no distant metastases, neutrophils ≥ 2000/uL, platelets ≥ 1000,000/uL, creatine < 130 umol/L, aminotransferases ≤ 3 x the upper limit of normal, and ≥ 18 years

Exclusion criteria: use of pilocarpine during RT and concomitant CT, second‐line treatment, incomplete assessment of salivary gland function

Age: intravenous: mean 55.2 range 34 to 78; subcutaneous: mean 56.1 range 36 to 76

Gender: intravenous: 127 M, 16F; subcutaneous: 124 M, 24 F

Cancer type: newly diagnosed squamous cell carcinoma of the head and neck, at all stages, and nodal status

Radiotherapy: at least 40 Gy of radiation delivered postoperatively

Chemotherapy: induction chemotherapy in 42 patients no concurrent chemotherapy

Number randomised: 291 (intravenous 143, subcutaneous 148)

Number evaluated: 127 (intravenous 67, subcutaneous 60) for xerostomia at 1 year

Interventions

Intravenous versus subcutaneous amifostine

Intravenous: 200 mg/m² daily, administered over 3 minutes, 15 to 30 minutes before RT

Subcutaneous: 500 mg at 2 sites, 20 to 60 minutes before RT

Outcomes

Xerostomia: grade 2 or above (0 to 4 scale). Physician graded via RTOG before treatment, every 3 months for the 1st year and then every 6 months

Salivary flow rates: unstimulated and stimulated saliva (mg/min)

Adverse effects: nausea, vomiting, hypotension, skin rash, local pain at injection site, fever, asthenia

Survival data: locoregional control, overall survival

Other oral symptoms: dysgeusia (taste disturbance), dysphagia (difficulty in swallowing), dysphonia (difficulty in speaking) ‐ these 3 items were combined with the patients' sensation of mouth dryness and assessed using a patient benefit questionnaire (see QoL); grade 3+ acute mucositis

Other oral signs: not reported

Quality of life: patient benefit questionnaire

Patient satisfaction: not reported

Cost data: not reported
Timing of assessment: acute xerostomia measured at 3 months; xerostomia, salivary flow rates and patient benefit questionnaire reported at 6 months, 1, 2 and 3 years; survival reported up to 4 years

Funding

Externally funded by pharmaceutical company; Schering‐Plough, France

Trial registration

clinicaltrials.gov/show/NCT00158691ID ‐ 12

Sample size calculation presented

Yes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Authors claim "randomly assigned". No further details given

Allocation concealment (selection bias)

Unclear risk

No details given

Blinding (performance bias and detection bias)
patients/carers

High risk

"Lack of double‐blind". Patients could not really be considered to be blinded as administration of amifostine differed

Blinding (performance bias and detection bias)
outcome assessment

High risk

Patient‐reported outcome see above

Incomplete outcome data (attrition bias)
All outcomes

High risk

Large loss to follow‐up. Attrition likely to be related to outcome

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Brizel 2000

Methods

Location: Europe, Canada, USA

Number of centres: 35 to 40 (unclear)

Date of enrolment: October 1995 to October 1997

33% dropout rate at 12 months

Participants

Inclusion criteria: patients with newly diagnosed, previously untreated squamous cell head and neck cancer. Inclusion of ≥ 75% of both parotid glands within radiation field and ≥ 40 Gy. Karnofsky Performance Status ≥ 60, granulocyte ≥ 2000 microL and platelet count ≥ 100,000 microL

Exclusion criteria: patients with T1N0 or T2N0 carcinomas of the true vocal cords and tumours of the major or minor salivary glands or history of malignancy other than in situ cervix carcinoma within 5 years preceding diagnosis. Pregnant women

Age: amifostine: 36 to 76, median = 55 years; control: 28 to 78, median = 56 years

Gender: amifostine 123 M, 27 F; control 120 M, 33 F

Cancer type: head and neck, various tumour sites, stages and node stages

Radiotherapy: amifostine: definitive = 50, postoperative high risk = 70 and postoperative low risk = 28; control: definitive = 52, postoperative high risk = 65 and postoperative low risk = 36. 1.8 to 2.0 Gy , 5 days a week over 5 to 7 weeks for a total dose of 50 to 70 Gy

Chemotherapy: none

Number randomised: 315 randomised, but 12 never received any treatment (amifostine 150, control 153)

Number evaluated: xerostomia at 12 months: 203 (amifostine 97, control 106), all included in analysis for locoregional control, all who received at least 1 dose of amifostine were assessed for toxicity

Interventions

Amifostine versus no intervention

Amifostine: (200 mg/m²) 3 minute intravenous 15‐30 minutes before RT
Control: nothing

Outcomes

Xerostomia: incidence of grade 2+ acute (within 90 days of the start of RT) and chronic xerostomia (0 to 4 scale)

Salivary flow rates: unstimulated and stimulated saliva production ‐ reported as median quantity (g) of saliva and also as number of participants producing > 0.1 g in 5 min ("a clinically relevant volume")

Adverse effects: nausea, vomiting, hypotension, allergic response

Survival data: locoregional control, progression‐free survival and overall survival at 24 months

Other oral symptoms: oral discomfort, dysgeusia (taste disturbance), dysphagia (difficulty in swallowing), dysphonia (difficulty in speaking) ‐ all included in patient benefit questionnaire (see QoL); grade 3+ acute mucositis

Other oral signs: not reported

Quality of life: patient benefit questionnaire (8 items each on a 10‐point scale where higher = better QoL)

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: xerostomia: within 3 months of start of RT, then at 12, 18 and 24 months; salivary flow rates: 12, 18 and 24 months after start of RT; quality of life: 12 months after start of RT

Funding

Source of funding: Medimmune Oncology

Trial registration

Not registered

Sample size calculation presented

Yes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "dynamic allocation process" (recognised methods referenced)

Allocation concealment (selection bias)

Low risk

Quote: "determined by a phone call from the enrolling institution to the protocol sponsor (US Bioscience)"

Comment: it appears to be central/remote allocation

Blinding (performance bias and detection bias)
patients/carers

High risk

Amifostine versus no intervention

Blinding (performance bias and detection bias)
outcome assessment

High risk

Open‐label, no blinded outcome assessment

Incomplete outcome data (attrition bias)
All outcomes

High risk

315 enrolled and randomised; 12 never received any treatment or follow‐up. Overall attrition 36%

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Brizel 2008

Methods

Location: Australia, Canada, USA

Number of centres: 22

Date of conduct: September 1999 to May 2001

Participants

Inclusion criteria: adults with newly diagnosed head and neck cancer. Patients with unknown primary and extensive neck disease also eligible. Karnofsky Performance Status > 60, haemoglobin > 10 g/dL, plus other similar criteria

Exclusion criteria: prior head and neck radiation therapy, prior surgery for primary tumour beyond biopsy, prior chemotherapy, known allergy to Escherichia coli‐derived products, participation in another study within the previous 30 days, refusal to use adequate contraception during study, pregnant or breastfeeding

Age: palifermin: mean 54 (range 25‐80); placebo: mean 56 (range 42‐75)

Gender: palifermin 55 M, 12 F; placebo 27 M 5 F

Cancer type: primary locations: oral cavity, oropharynx/nasopharynx, hypopharynx/larynx

Radiotherapy: isocentric 4 to 6 MV photons either standard fractionation (once daily 2 Gy fractions 5 days/week: total primary tumour dose 70 Gy) or hyperfractionation (single 2 Gy fraction followed by a planned 1‐week treatment break. Then twice‐daily radiation: total dose of 72 Gy/6.5 weeks). Varied by centre
Chemotherapy: cisplatin 20 mg/m² per day as IV bolus injection and fluorouracil 1000 mg/m² per day as continuous infusion, both on 1st 4 days of 1st and 5th weeks of RT

Number randomised: 101 (69 palifermin, 32 placebo)

Number evaluated: varies by outcome but 97 (65 palifermin, 32 placebo) analysed for our primary outcome of xerostomia

Interventions

Palifermin versus placebo

Palifermin: 60 µg/kg by IV bolus injection on study day 1 (Friday) before 1st week of CRT. Subsequent doses administered for 7 consecutive weeks, on each Friday after completion of weekly radiation treatment. 2 additional doses given on weeks 8 and 9

Placebo: as above

Follow‐up: 5 weeks after end of RT

Outcomes

Xerostomia: incidence of grade 2 xerostomia using NCI CTC scale

Salivary flow rates: not reported

Adverse effects: nausea, vomiting, fever, constipation, dehydration, granulocytopenia, fatigue, diarrhoea, insomnia, anaemia, dysaphia, cough, headache, weight decrease, dizziness, anxiety, hypomagnesaemia

Survival data: survival, progression‐free survival (up to 75 months)

Other oral symptoms: mucositis (primary outcome of study), dysphagia

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Funding

Pharmaceutical trial (Amgen)

Trial registration

Not registered

Sample size calculation presented

Yes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

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

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding (performance bias and detection bias)
patients/carers

Low risk

Quote: "double‐blind randomised placebo‐controlled study"

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Quote: "double‐blind randomised placebo‐controlled study". However there is a subjective element to the index

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Xerostomia data on 97 out of 101 enrolled. Quote: "3 patients in palifermin group and 1 in the placebo group discontinued study treatment with adverse events not considered related to study treatment". Comment: unclear if 3 of these were missing for xerostomia

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Buentzel 2006

Methods

Location: Europe, USA

Number of centres: 18 (15 Europe, 3 USA)

Date of recruitment: October 1996 to October 1998

Participants

Inclusion criteria: at least 18 years of age scheduled for definitive or adjuvant chemoradiotherapy for histologically confirmed squamous cell carcinoma of the head and neck. Postsurgery the surgical wound must be healed but no later that 12 weeks after surgery. Inclusion of at least 75% of each parotid gland within radiation field. Life expectancy 12+ months, Karnofsky Performance Status 60+, adequate function of bone marrow, kidneys and the liver

Exclusion criteria: evidence of distant metastatic disease, primary lesion of the parotid gland, or a history of prior malignancy within the past 5 years (other than non‐melanomatous skin cancers that are controlled or carcinoma in situ of the cervix). Scheduled to receive hyperfractionated or accelerated radiotherapy, previously treated with chemotherapy or other investigational therapies within 4 weeks of study entry. Pregnant women

Age: amifostine: median 57 (range 29‐73); placebo: median 58 (range 23‐78)

Gender: amifostine 54 M, 13 F; placebo 57 M, 8 F

Cancer type: head and neck cancer, various primary sites and stages

Radiotherapy: standard fractionation (1.8‐2.0 Gy per day, 5 days a week) over 6 to 7 weeks for a total dose of 60 to 70 Gy

Chemotherapy: carboplatin 70 mg/m² IV over 30 minutes after amifostine and 30 minutes before RT

Number enrolled: 132

Number randomised: 132

Number evaluated: 132 (ITT analysis) (67 amifostine; 65 placebo)

Interventions

Amifostine versus placebo

Amifostine: 300 mg/m² IV over 3 minutes (days 1‐5 and 21‐25 of treatment); 200 mg/m² IV over 3 minutes (days 6‐20 and 26‐30/35)
Placebo (Mannitol): equivalent volume to amifostine

Outcomes

Xerostomia: RTOG acute and late radiation morbidity scoring criteria; incidence of grade 2 or higher acute or late xerostomia (0 to 4 scale)

Salivary flow rates: stimulated and unstimulated saliva measurements (not assessed as less than a 3rd of participants had salivary function at 1 year)

Adverse effects: nausea, vomiting, allergic response, asthenia

Survival data: locoregional failure rate, progression‐free survival and overall survival

Other oral symptoms: RTOG acute and late radiation morbidity scoring criteria: grade 3 or higher acute mucositis

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: acute xerostomia and mucositis measured up to 90 days after start of RT; late xerostomia measured up to 12 months after start of RT

Funding

Source of funding: MedImmune Oncology Inc grant

Trial registration

Not registered

Sample size calculation presented

Yes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Assumed. Described as "dynamic allocation scheme", similar method to Brizel 2000

Allocation concealment (selection bias)

Low risk

Fax of baseline data sent to central telephone number for randomisation number. Randomisation number identical to blinded drug container held at pharmacy

Blinding (performance bias and detection bias)
patients/carers

Low risk

Amifostine versus placebo

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Information provided by author: "blinded drug containers were kept at the pharmacy, treating physicians had no information about the randomization until the end of the follow‐up period"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Analyses carried out an ITT basis. Dropouts = 30 (23% dropout rate). Amifostine group = 21 (1 ‐ never treated, 16 ‐ toxicity, 1 ‐ patient request, 1 ‐ death, 2 ‐ other illness). Placebo group = 10 (1 ‐ never treated, 4 ‐ toxicity, 1 ‐ patient request, 1 ‐ death, 1 ‐ disease progression, 2 ‐ non‐compliance)

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Burlage 2008

Methods

Location: the Netherlands

Number of centres: 2

Date of enrolment: April 1999 ‐ October 2003

Participants

Inclusion criteria: biopsy confirmed HNSCC, initial 5% (wt/vol) citric acid‐stimulated parotid salivary flow > 0.1 mL/min

Exclusion criteria: previous irradiation and/or previous or concurrent chemotherapy, patients with salivary gland tumours, severe cardiovascular disease or chronic obstructive pulmonary disease, pregnant women

Age: pilocarpine: 18‐60 years 50 participants, > 60 years 35 participants; placebo: 18‐60 years 42 participants, > 60 years 42 participants

Gender (M:F): pilocarpine 22:63; placebo 13:71

Cancer type: oral cavity (17%), oropharynx (18%), larynx (51%), hypopharynx (7%), nasopharynx (4%), unknown primary (1%) (equally distributed across groups)
Submandibular gland removal: both removed: pilocarpine 2%, placebo 5%; 1 removed: pilocarpine 37%, placebo 38%

Radiotherapy: clinical target volume of initial field encompassed the primary tumour site with 1.5 cm margin, neck node levels in which pathologic nodes were found and elective node areas on both sides. Conventional fractionation schedule. Received at least 40 Gy in daily 2 Gy fractions

Chemotherapy: none

Number randomised: 170 (85 per group)

Number evaluated: 113 (pilocarpine 55, placebo 58)

Interventions

Pilocarpine versus placebo

Pilocarpine: 5 mg 4 times daily 2 days before start of RT until 14 days after RT

Placebo: similar tablets, same schedule

Outcomes

Xerostomia: from validated head‐and‐neck symptom questionnaire on 5‐point scale; LENT SOMA

Salivary flow rates: parotid salivary flow using Carlson‐Crittenden cups, from left and right hand parotid glands simultaneously under standardised conditions for 10 min. Flow stimulated with 5% (wt/vol) citric acid. Parotid flow complication probability also reported

Adverse effects: not reported

Survival data: locoregional control

Other oral symptoms: eating, swallowing

Other oral signs: not reported

Quality of life: some covered in validated head‐and‐neck symptom questionnaire on 5‐point scale

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: before RT, 6 weeks, 6 months and 12 months postRT

Funding

Not reported. Conflicts of interest: "none" reported

Trial registration

Not registered

Sample size calculation presented

Yes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomisation was executed by the hospital pharmacist by computer, using random permuted blocks within strata. The randomisation key was opened after the last saliva collection (1 year after the last patient was included and after completion of all planned assessments)"

Allocation concealment (selection bias)

Low risk

See above

Blinding (performance bias and detection bias)
patients/carers

Low risk

Quote: "Double‐blind randomised placebo‐controlled study". Intervention was tablets supplied by the pharmacy

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Quote: "Double‐blind randomised placebo‐controlled study". Intervention was tablets supplied by the pharmacy

Incomplete outcome data (attrition bias)
All outcomes

High risk

32% missing at 12 months with no clear reasons given by study group

Selective reporting (reporting bias)

High risk

Adverse events and xerostomia data not fully reported

Other bias

Low risk

No other sources of bias are apparent

Büntzel 1998

Methods

Location: Germany

Number of centres: 1

Date of recruitment: not stated

Participants

Inclusion criteria: stage III or IV carcinoma of the head and neck, aged 16 to 80 and no evidence of systemic infection or liver or renal impairment. Tumour resected or excised before adjuvant RT

Exclusion criteria: not reported

Age: amifostine: median 61 (range 40‐77); control: median 58 (range 38‐75)

Gender: amifostine 13 M, 1 F; control 12 M, 2 F

Cancer type: tumour location (amifostine/control): larynx = 3/1, hypopharynx = 4/3, mesopharynx = 3/7, nose = 2/1, mouth = 2/2

Radiotherapy: 2 Gy fractions, 5 days a week for 6 weeks; maximum dose of 60 Gy (encompassing 75% of the major salivary glands)

Chemotherapy: 20 min IV infusion of carboplatin (70 mg/m² days 1 to 5 and 21 to 25 of treatment)

Number randomised: 28 (14 amifostine, 14 control)

Number evaluated: 28

Interventions

Amifostine versus no intervention

Amifostine: (500 mg) 15 min IV before carboplatin (days 1 to 5 and days 21 to 25). Followed by antiemetic regimen to control nausea/vomiting
Control: nothing

Use of supportive drugs reported: (amifostine/control): G‐CSF: 2/7; GM‐CSF: 0/7; antibiotics: 4/10

Outcomes

Xerostomia: incidence and severity using WHO grading (0 to 4 scale ‐ we report grade 2 and above)

Salivary flow rates: not reported

Adverse effects: hypotension

Survival data: not reported

Other oral symptoms: dysgeusia (taste disturbance), dysphagia (difficulty in swallowing), mucositis (WHO)

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: economic evaluation (Bennett 2001)

Timing of assessment: xerostomia at end of RT and 1 year; other oral symptoms at end of RT

Funding

US Bioscience who produce Ethyol‐amifostine

Trial registration

Not registered

Sample size calculation presented

Not reported

Notes

Additional data presented but included extra 11 patients in amifostine group who were not entered in the study (not included in analyses)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding (performance bias and detection bias)
patients/carers

High risk

Amifostine versus no intervention

Blinding (performance bias and detection bias)
outcome assessment

High risk

Not blinded and xerostomia is a subjective outcome

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants included in analysis

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Büntzel 2010

Methods

Location: Germany

Number of centres: 6

Date of study: 2001 to 2007

Participants

Inclusion criteria: SCCHN with deficiency in selenium and if radiation field included 75% of the major salivary glands

Exclusion criteria: none reported

Age: median 63.2 range 38.7‐83.0

Gender (M:F): selenium 16:6; control 15:2

Cancer type: head and neck cancer

Radiotherapy: 1.8 to 2.0 Gy to primary tumour and lymphatic neck during daily radiation treatment; to total dose 60‐72 Gy

Chemotherapy: unclear

Number randomised: 40: 22 selenium, 18 control

Number evaluated: 39: 22 selenium, 17 control

Interventions

Selenium versus no intervention

Selenium: 500 µg sodium selenite, 2 days before RT, 500 µg selenite and radiation days (300 µg if official holiday). Administrated as oral fluid 1 hour before RT

Control: no intervention

Outcomes

Xerostomia: RTOG grade for xerostomia

Salivary flow rates: not reported

Adverse effects: serious adverse events reported

Survival data: not reported

Other oral symptoms: mucositis RTOG, dysgeusia (taste disturbance RTOG), dysphagia (difficulty in swallowing RTOG)

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: 1, 2, 3, 4, 5, 6, 7 weeks from start of RT and 6 weeks after RT

Funding

Externally funded by Arzneimittel, Germany

Trial registration

Unclear

Sample size calculation presented

Not reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not given, stated "randomised"

Allocation concealment (selection bias)

Unclear risk

Allocation after consent obtained

Blinding (performance bias and detection bias)
patients/carers

High risk

Participants receive selenium oral fluid prior to radiotherapy or not. Blinding not possible

Blinding (performance bias and detection bias)
outcome assessment

High risk

Participants receive selenium oral fluid prior to radiotherapy or not, and their subjective assessment of xerostomia is included

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Initial study requiring 60 patients per arm stopped early due to slow accrual

113 screened. 93 selenium deficient. 40 consented. 1 withdrawal, 39 reported

Selenium concentrations reported in other article elsewhere

Selective reporting (reporting bias)

High risk

Xerostomia but no standard deviations. Total adverse events reported but not per person

Other bias

Low risk

No other sources of bias are apparent

Duncan 2005

Methods

Location: Canada

Number of centres: multicentre (unclear how many)

Date of enrolment: September 1997 to September 1999

Participants

Inclusion criteria: SCCHN, non‐metastatic disease

Exclusion criteria: none reported

Age: lozenge median 59.7; placebo median 57.3

Gender (M:F) : lozenge (48:18), placebo (52:15)

Cancer type: oral cavity, oropharynx, hypopharynx, nasopharynx, larynx

Radiotherapy: conventional radical or postoperative radiotherapy to a dose of 50 Gy or greater delivered in once daily fractions (1.8 to 2.4 Gy)

Chemotherapy: not mentioned, probably none

Number randomised: 138 (69 per group)

Number evaluated: 133 (lozenge 66; placebo 67)

Interventions

Antimicrobial lozenge versus placebo

Antimicrobial lozenge: BCoC, bacitracin, 6 mg; clotrimazole, 10 mg; gentamicin, 4 mg. Unclear how frequently taken or for how long

Placebo: not described ‐ assumed similar

Outcomes

Xerostomia: item on trial specific checklist ‐ 'Did you have mouth dryness (1‐4 scale)?' and NCIC CTG ECTC physician‐rated (using patient diary)

Salivary flow rates: not reported

Adverse effects: not reported

Survival data: not reported

Other oral symptoms: mucositis using OMAS (primary outcome), mouth pain, chewing, numbness, mouth opening, burning mouth

Other oral signs: not reported

Quality of life: 2 tools ‐ European Organisation for Research and Treatment of Cancer Quality of Life questionnaire (EORT QLQ‐C30), trial specific checklist

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: 2, 4, 6 during RT; 8‐9, 12‐14, 24 weeks on study

Funding

The National Cancer Institute of Canada, Clinical Trials Group

Trial registration

Unclear

Sample size calculation presented

No

Notes

Primarily study to prevent mucositis

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomly assigned"

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding (performance bias and detection bias)
patients/carers

Low risk

Quote: "double‐blind controlled trial". Placebo tablets given

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Quote: "double‐blind controlled trial". Placebo tablets given

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Compliance with quality of life forms reported to be 93.3% but reasons for dropouts not reported. Similar low rates of attrition per group

Selective reporting (reporting bias)

High risk

Xerostomia reported, adverse events not reported for lozenge

Other bias

Low risk

No other sources of bias are apparent

Fisher 2003

Methods

Location: USA

Number of centres: unclear

Date of randomisation: March 1998 to February 2000

Participants

Inclusion criteria: oral and oropharyngeal squamous cell carcinoma, Karnofsky Performance Score ≥ 60, no prior radiotherapy to the head and neck, planned irradiation of the oral cavity or oropharynx in which at least 50% of the major salivary glands are to receive > 50 Gy

Exclusion criteria: salivary gland malignancy; use of cholinergic, anticholinergic, and tricyclic drugs; and patients with uncontrolled asthma, acute iritis, or narrow‐angle glaucoma

Age: pilocarpine 60.8 years; placebo 59.3 years

Gender (M:F): pilocarpine 93:28, placebo 92:32

Cancer type: oral cavity 52; nasopharynx 3; oropharynx 104; hypopharynx 11; other 13; unknown 18 (evenly distributed across groups)

Radiotherapy: 60‐70 Gy with 50% of volume of major salivary glands receiving 50 Gy

Chemotherapy: not stated

Number randomised: 249; 3 ineligible, all from pilocarpine arm (121 pilocarpine, 125 placebo)

Number evaluated: 166 end of RT (pilocarpine 89, placebo 77); 166 at 3 months (pilocarpine 85, placebo 81); 137 at 6 months (pilocarpine 68, placebo 69)

Interventions

Pilocarpine versus placebo

Pilocarpine: 5 mg tablets 4 times daily starting 3 days before RT and continuing for 3 months
Placebo: 5 mg tablets 4 times daily starting 3 days before RT and continuing for 3 months. 3 months after RT the placebo group were permitted to cross over to pilocarpine

Outcomes

Xerostomia: not reported

Salivary flow rates: salivary gland scintigraphy (stimulated and unstimulated)

Adverse effects: drug toxicities reported

Survival data: not reported

Other oral symptoms: RTOG acute mucositis, mouth pain, dysgeusia (taste disturbance), dysmasesia (difficulty in chewing), dysphagia (difficulty in swallowing), dysphonia (difficulty in speaking)

Other oral signs: not reported

Quality of life: University of Washington QoL scale

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: pretreatment, end of RT, 3 months after end of RT, 6 months after end of RT

Funding

National Cancer Institute and MGI Pharma Inc.

Trial registration

clinicaltrials.gov/show/NCT00003139ID ‐ 11

Protocol available

Sample size calculation presented

Not reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding (performance bias and detection bias)
patients/carers

Low risk

Pilocarpine versus placebo

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Pilocarpine versus placebo

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

213/249 available for analysis. Dropouts very high for salivary flow (33% end of RT and 3 months, 45% at 6 months)

Selective reporting (reporting bias)

High risk

Xerostomia not reported

Other bias

Low risk

No other sources of bias are apparent

Gornitsky 2004

Methods

Location: Canada

Number of centres: 1

Date of randomisation: March 1998 to September 2001

Participants

Inclusion criteria: scheduled to receive external beam radiotherapy, using a bilateral radiation technique encompassing ≥ 2/3 of all major and minor salivary glands for a minimum of 5000 cGy (200 cGy per day) for 5‐7 weeks

Exclusion criteria: clinically significant cardiovascular disease, chronic obstructive pulmonary disease, biliary tract disease, uncontrolled asthma, acute iritis, narrow angle glaucoma, participants who are pregnant or nursing. Hypersensitivity to pilocarpine, participants on tricyclic antidepressants, antihistamines with anticholinergic effects, beta blockers, or pilocarpine for ophthalmic indications were excluded

Age (mean): pilocarpine 58 years; placebo 61 years

Gender (M:F): pilocarpine 26:3, placebo 24:5

Cancer type: oral cavity 14; pharynx 13; tonsil 11; glottis 3; larynx 11; sinus 2; neck 1; unknown 1 (evenly distributed across groups)

Radiotherapy: Mean dose = 64.7 Gy (pilocarpine group), 63.7 Gy (placebo group)

Chemotherapy: pilocarpine 13 (45%); placebo 9 (32%)

Number randomised: 58

Number evaluated: 58 (22 dropped out but ITT was used and missing data were calculated)

Interventions

Pilocarpine versus placebo

Phase 1
Pilocarpine: 5 mg tablets 5 times daily, half an hour before meals, before radiotherapy, and prior to sleep during the period of radiotherapy

Placebo: identical tablets 5 times daily, half an hour before meals, before radiotherapy, and prior to sleep during the period of radiotherapy

Phase 2
All received pilocarpine (5 mg) 4 times daily half an hour before meals and prior to sleep for 5 weeks

Outcomes

Xerostomia: subjective assessment of xerostomia: VAS (rated 0‐100)

Salivary flow rates: whole saliva secretion (unstimulated and stimulated) using the SAXON test

Adverse effects: not reported (data provided by author)

Survival data: not reported

Other oral symptoms: oral discomfort, difficulty with eating, dysphonia (difficulty in speaking), mucosal pain or burning (VAS, rated 0‐100)

Other oral signs: not reported

Quality of life: global quality of life, sleeping problems (VAS, rated 0‐100)

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: prior to RT, end of RT, 5 weeks after end of RT

Funding

Pharmacia Canada

Trial registration

Not registered

Sample size calculation presented

Not reported

Notes

Phase 2 data not included in the review

Additional data provided by author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Comment from author: "block of four using a random number table... allocation sequence prepared by pharmacy of Jewish General Hospital"

Allocation concealment (selection bias)

Low risk

Third party randomisation; coded bottles

Blinding (performance bias and detection bias)
patients/carers

Low risk

Bottles only distinguished by number allocated by pharmacy. Investigators, treating physicians and patients blinded

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Subjective outcomes self reported (patients unaware of treatment group)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Analysis carried out on ITT basis. 38% dropout rate. 58 randomised, 22 dropped out (9 pilocarpine, 13 placebo)

Selective reporting (reporting bias)

High risk

Adverse events not reported

Other bias

Low risk

No other sources of bias are apparent

Grötz 2001

Methods

Location: Germany

Number of centres: 1

Date of randomisation: not stated

Participants

Inclusion criteria: scheduled to receive adjuvant or sole radiotherapy for head and neck cancer to a scheduled dose of 60 Gy. Cranial border of the field above the chin‐mastoid line so salivary glands are located in the core irradiation field

Exclusion criteria: salivary gland disorders

Age: mean age = 55 years

Gender: 22 M, 1 F

Cancer type: head and neck

Radiotherapy: total dose = 60 Gy
Chemotherapy: unclear

Number randomised: 48

Number evaluated: 23

Interventions

Coumarin + troxerutin versus placebo

Venalot Depot (coumarin 15 mg and troxerutin 90 mg) tablet: 2 tablets 3 times daily. Start 1 week before RT and 4 weeks after end of RT
Control: placebo

Outcomes

Xerostomia: not reported, only as part of total RTOG

Salivary flow rates: stimulated and unstimulated using sialoscintigraphy (sialometry abandoned as primary marker as not successfully collected). Acute radiation side effects RTOG score but for all organs

Adverse effects: reddened skin, nausea

Survival data: locoregional control

Other oral symptoms: not reported

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: 4 weeks after RT

Funding

Not stated

Trial registration

Unclear

Sample size calculation presented

Not reported

Notes

Unable to use data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding (performance bias and detection bias)
patients/carers

Low risk

Venalot Depot versus placebo

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Salivary flow rates objective outcome

Incomplete outcome data (attrition bias)
All outcomes

High risk

48 randomised, 25 dropped out. Dropouts per group not specified

Selective reporting (reporting bias)

High risk

Xerostomia not reported. Data for total RTOG score presented but no break down by condition or organ

Other bias

Low risk

No other sources of bias are apparent

Haddad 2002

Methods

Location: Iran

Number of centres: 1

Date of recruitment: 1998‐2000

Participants

Inclusion criteria: 18‐70 year old patients, irradiated to the head and neck, both parotid glands in the radiation fields (minimum 40 Gy). No previous history of irradiation in this region
Exclusion criteria: asthma, chronic obstructive pulmonary disease, narrow‐angle glaucoma, biliary or renal lithiasis and hypertensive, heart or psychiatric disorders requiring medical treatment

Age: mean across groups = 43 years (range 18 to 70 years)

Gender (M:F): across groups 36:24

Cancer type: primary site of tumour. Pilocarpine group: nasopharynx (n = 17), neck adenopathy (n = 1). Placebo group: maxilla (n = 2), nasopharynx (n = 13), tongue (n = 1), tonsil (n = 5)

Radiotherapy: standard fractionation (1.8 to 2 Gy per day, 5 days a week) and cobalt‐60 systems; mean parotid dose 58 Gy (pilocarpine 59 Gy; placebo 57 Gy) (range 45 to 70 Gy)
Chemotherapy: none

Number randomised: 60

Number evaluated: 39 (18 pilocarpine, 21 placebo)

Interventions

Pilocarpine versus placebo

Pilocarpine hydrochloride: 5 mg 3 times daily for 3 months starting from the beginning of RT
Placebo: 5 mg 3 times daily for 3 months starting from the beginning of RT

Outcomes

Xerostomia: subjective evaluation score for xerostomia using 6 questions evaluated using VAS (0‐100 mm). Objective grading of xerostomia according to the Late Effects of Normal Tissues Subjective, Objective, Management and Analytic (LENT SOMA) scale

Salivary flow rates: not reported

Adverse effects: lacrimation (excess tears, crying), nausea

Survival data: overall survival

Other oral symptoms: not reported

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: 6 months postRT

Funding

Source of funding: Tehran University of Medical Sciences' research grant

Trial registration

Not registered

Sample size calculation presented

Not reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "...randomisation was performed at the start of radiotherapy by the sealed envelope method"
Comment: although not clear the randomisation was probably done well as the pharmacy was involved in making and distributing the tables

Allocation concealment (selection bias)

Low risk

Sealed envelopes, pharmacy involvement

Blinding (performance bias and detection bias)
patients/carers

Low risk

Capsules only distinguished by a number recorded by the drug manufacturer. Investigators, treating physicians and patients blinded

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Capsules only distinguished by a number recorded by the drug manufacturer. Investigators, treating physicians and patients blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

60 enrolled. 13/31 (42%) dropouts in pilocarpine group; 8/29 (28%) dropouts in placebo group

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Haddad 2009

Methods

Location: USA

Number of centres: 4

Date of enrolment: May 2003 to April 2006

Participants

Inclusion criteria: with stage III or IV, previously untreated, locally advanced, SCCHN. Primary tumour types allowed: oropharynx, hyperpharynx, oral cavity, larynx, unknown primary

Exclusion criteria: grade > 2 peripheral neuropathy other serious comorbid illness, involuntary weight loss of > 20% of body weight in 3 months preceding study

Age: amifostine mean 55; control 57

Gender: amifostine: 27 M, 2 F; control: 23 M, 6 F

Cancer type: (amifostine/control) oropharynx = 18/17, oral cavity = 5/6, larynx = 3/5, unknown primary = 2/0, other = 1/1
Neck dissection: amifostine 48%; control 38%; no details reported

Radiotherapy: concomitant boost radiation, 72 Gy in 42 fractions over 6 weeks. Use of IMRT not allowed

Chemotherapy: 4 weekly doses of carboplatin/paclitaxel. Induction chemotherapy was used in 29 of 58 patients overall with docetaxel, cisplatin, and 5‐fluorouracil

Number randomised: 58 (29 per group)

Number evaluated: unclear for xerostomia

Interventions

Amifostine versus no intervention

Subcutaneous daily amifostine at dose of 500 mg 30‐60 min before daily RT (before morning dose only, when schedule moved to twice daily radiotherapy at day 19). Average number of amifostine doses was 25 (median 28 doses). Amifostine withheld for skin toxicity

Outcomes

Xerostomia: Common Terminology Criteria for Adverse Events including xerostomia reported but not by group

Salivary flow rates: saliva collection with and without citric acid simulation

Adverse effects: not reported

Survival data: overall survival, progression‐free survival, local control

Other oral symptoms: Common Terminology Criteria for Adverse Events for mucositis, swallowing measured

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: xerostomia and mucositis assessed weekly throughout RT, then every 4 weeks after RT; salivary flow rate assessed at 12, 24 and 52 weeks after RT; dysphagia (swallowing) assessed at 8, 12, 24 and 52 weeks after RT; survival ‐ median follow‐up 34 months after RT, minimum 26 months

Funding

Medimmune Oncology

Trial registration

Not registered

Sample size calculation presented

Yes

Notes

Quote: "Study stopped before completion of planned accrual because IMRT was becoming de facto standard technique in treating head and neck cancer"

Study focuses on survival

Not able to use data ‐ contacted authors for data 19 February 2016

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The randomisation process was centralised and managed through the Dana‐Farber Cancer Institute protocol office" Comment: linked to Harvard University probably done well

Allocation concealment (selection bias)

Low risk

Quote: "The randomisation process was centralised and managed through the Dana‐Farber Cancer Institute protocol office"

Blinding (performance bias and detection bias)
patients/carers

High risk

No intervention group as comparator ‐ not blinded

Blinding (performance bias and detection bias)
outcome assessment

High risk

Subjective assessment of xerostomia

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear how many participants dropped out

Selective reporting (reporting bias)

High risk

Badly reported xerostomia and no adverse events

Other bias

Low risk

No other sources of bias are apparent

Han 2010

Methods

Location: China

Number of centres: 2

Date of conduct: 1 October 2007 to 31 July 2009

Participants

Inclusion criteria: quote: "First‐visit patients; diagnosed as mid/moderate to advanced/terminal nasopharyngeal squamous carcinoma through pathological and radiographic examinations; Karnosfsky score ≥ 60; expected survival period > 6 months; without severe complications (e.g. hypertension, coronary heart disease, diabetes, history of mental illness)"

Exclusion criteria: see above

Age: Jinlong: mean 46.3 (SD 7.4), median 53; control: mean 47.4 (SD 6.8), median 52

Gender: Jinlong: 33 M, 16 F; control: 34 M, 14 F

Cancer type: nasopharyngeal squamous carcinoma

Radiotherapy: dose 60 to 76 Gy, 2 Gy per day, 5 times a week

Chemotherapy: "concurrent chemoradiotherapy" (no further details)

Number randomised: 97 (Jinlong: 49, control: 48)

Number evaluated: 95 (Jinlong: 48, control: 47)

Interventions

Jinlong capsules versus no intervention

4 tablets once, 3 tablets every day

Duration: 3 months

Follow‐up: 12 weeks after treatment

Quote: "Jinlong capsule is a modern 'fresh medicine preparation' made of fresh gecko and fresh long‐noded pit vipers, using cryogenic modern biochemical extracting and separation techniques. It maintained to the greatest degree the activity of effective ingredients of organisms, and reasonable compatibility among the ingredients. Basic research has shown that Jinlong can directly damage cancer cells by blocking the mitosis and proliferation of cancer cells, fix the p21 small protein molecule, restore the regulation of cancer cells, and turn cancer cells to normal cells..."

Outcomes

Xerostomia: quote: "observe the patients for toxic and side effects during and after radiotherapy, assess the toxic and side effects according to RTOG's criteria"

Salivary flow rates: not reported

Adverse effects: leukopenia, nausea, vomiting, 1 participant had dizziness and blood pressure drop, 1 participant had skin rash

Survival data: not reported

Other oral symptoms: mucositis

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Funding

Not reported; conflicts of interest: not reported

Trial registration

Not registered

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomly divided"

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding (performance bias and detection bias)
patients/carers

High risk

Jinlong versus no intervention

Blinding (performance bias and detection bias)
outcome assessment

High risk

Jinlong versus no intervention

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quotes: "1 patient quit because of myocardial infarction (Tx Group)", "1 patient quit because of mucosa toxicity (control group)"

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

He 2004

Methods

Location: China

Number of centres: 1

Date of conduct: not stated

Participants

Inclusion criteria: aged 20‐70 years; Karnofsky Performance Score > 70; Hb 90 to 150/L; blood pressure 12‐20/8‐15 kPa; normal kidney and liver function; no severe infection such as septicaemia; no heart disease; no medical history of low blood pressure, no other cancer and no history of radiotherapy

Exclusion criteria: see above

Age: aged 20 to 70 (no further details)

Gender: not reported

Cancer type: amifostine: nasopharyngeal squamous cell carcinoma stage 1 = 1, stage 2 = 7, stage 3 = 8 and stage 4 = 1. Control: nasopharyngeal squamous cell carcinoma stage 1 = 1, stage 2 = 5, stage 3 = 1 and stage 4 = 1

Radiotherapy: conventional with nasopharyngeal tumour dose (65‐74 Gy)
Chemotherapy: none

Number randomised: 32 (amifostine: 17; control: 15)

Number evaluated: 32 (amifostine: 17; control: 15) ‐ 1 participant left amifostine group due to GI tract side effect but analysis states 17 in this group (possible ITT analysis)

Interventions

Amifostine versus no intervention

Amifostine (200 mg/m²), diluted with 'water for injection' at the concentration of 50 mg/mL, IV 15‐30 min before RT
Control: nothing

Outcomes

Xerostomia: "mucositis and xerostomia according to RTOG's criteria" (0‐4 scale; we report grade 2 and above)

Salivary flow rates: "method used to measure the amount of saliva: put a 0.2 g cotton ball under patient's tongue, after 3 minutes, use electronic balance to measure its weight", reported as decrease in saliva/change score (unstimulated)

Adverse effects: GI tract reaction/side effects (nausea and vomiting)

Survival data: not reported

Other oral symptoms: mucositis (RTOG criteria)

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: 3, 5 and 7 weeks after start of RT

Funding

Not reported; conflicts of interest: not reported

Trial registration

Not registered

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomized into"

Comment: no further details given

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding (performance bias and detection bias)
patients/carers

High risk

Amifostine versus no intervention

Blinding (performance bias and detection bias)
outcome assessment

High risk

Not possible due to no intervention group and subjective assessment of xerostomia

Incomplete outcome data (attrition bias)
All outcomes

Low risk

1/32 participants dropped out, however, appears to be included in analysis

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Henke 2011

Methods

Location: Australia, Canada and Europe

Number of centres: 38 hospitals

Date of conduct: January 2005 to August 2007

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

Age: palifermin: mean 56 (SD 8); placebo: mean 57 (SD 9)

Gender: palifermin: 78 M, 14 F; placebo: 75 M, 19 F

Cancer type: head and neck (oropharynx, oral cavity, larynx, hypopharynx, other)

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) IV after appropriate hydration on days 1 and 22 (for R0 resection), or days 1, 22 and 43 (for R1 resection)

Number randomised: 186 (palifermin 92; placebo 94)

Number evaluated: 186 (palifermin 92; placebo 94)

Interventions

Palifermin versus placebo

Palifermin: (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 or 960 µg/kg respectively)

Placebo: same schedule with placebo

Outcomes

Xerostomia: incidence of grade ≥ 2 xerostomia (Common Terminology Criteria for Adverse Events (CTCAE) v 3.0, assessed at months 4, 6, 8, 10, 12, reported only at month 4

Salivary flow rates: not reported

Adverse effects: assessed weekly during study treatment

Survival data: overall and progression‐free survival, incidence of disease recurrence and death

Other oral symptoms: incidence of dysphagia (difficulty in swallowing), OMWQ‐HN 0 (no soreness) to 4 (extreme soreness) scale for mouth and throat soreness assessed weekly and reported as mean score

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Funding

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

Trial registration

NCT00131638 (clinicaltrials.gov/ct2/show/NCT00131638)

Sample size calculation presented

Yes

Notes

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 (performance bias and detection bias)
patients/carers

Low risk

Quote: "placebo‐controlled, double‐blind study"
Comment: blinding feasible

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Quote: "placebo‐controlled, double‐blind study"
Comment: blinding feasible

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All cases accounted for. ITT analysis (participants having no assessment assumed to have event)

Selective reporting (reporting bias)

Low risk

All outcomes reported. Low risk for xerostomia at the time point used in meta‐analysis (4 months) ‐ however, it should be noted that xerostomia was measured up to 12 months but data not reported

Other bias

Low risk

No other sources of bias are apparent

Hu 2005

Methods

Location: China

Number of centres: 1

Date of conduct: January 2002 to June 2004

Participants

Inclusion criteria: head and neck patients confirmed by pathological examination

Exclusion criteria: not reported

Age (years): treatment: mean 51 (SD 19); control: mean 49 (SD 18)

Gender: treatment: 36 M 34 F; control: 38 M, 32 F

Cancer type: treatment: nasopharyngeal (52), tonsil (11) and tongue (7); cancer stage: I = 6, II = 20, III = 28 and IV = 16. Control: nasopharyngeal (51), tonsil (11) and tongue (8); cancer stage: I = 6, II = 19, III = 29 and IV = 16

Radiotherapy: overall dose: 70 Gy for nasopharyngeal carcinoma, 55‐70 Gy for carcinoma of tonsil and tongue

Chemotherapy: none

Number randomised: 140 (treatment 70, control 70)

Number evaluated: 140 (treatment 70, control 70)

Interventions

Shenqi Fanghou recipe versus no intervention

Shenqi Fanghou recipe: dangshen (30 g), astragalus root (30 g), tuckahoe (30 g), Chinese yam (30 g), hedyotic diffusa (30 g), barbated skullcup herb (30 g), pueraria root (30 g), fragrant solomonseal rhizome (10 g), glossy privet fruit (10 g), stiff silkorm (10 g), grassleaf sweetflag rhizome (10 g), atractylodes macrocephala (10 g), semen coicis (50 g), dried tangerine peel (6 g), paris root (20 g), figwort root (15 g), common anemarrhena rhizome (15 g), gambir plant (15 g), scorpion (5 g), radix notoginseng (5 g), radix glycyrrhizae (5 g)
Dosage: solution of 400 ml (200 ml in the morning and 200 ml in the afternoon), starting the first day of RT for 35 to 38 days
Control group: nothing

Follow‐up: end of RT

Outcomes

Xerostomia: subjective assessment of dry mouth: 1) mild: can eat dry cooked rice, 2) moderate: have difficulty in eating dry cooked rice, or 3) severe: cannot eat dry cooked rice

Salivary flow rates: not reported

Adverse effects: none

Survival data: survival after a follow‐up of more than 1 year

Other oral symptoms: oropharyngeal mucosa reaction, difficulty in mouth opening

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Funding

Source of funding: government (The Bureau of Science and Technology of Shenzhen City); conflicts of interest: not reported

Trial registration

Not registered

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomly divided"

Comment: no further details given

Allocation concealment (selection bias)

Unclear risk

Quote: "the envelop method was used to randomise"

Comment: insufficient information

Blinding (performance bias and detection bias)
patients/carers

High risk

Shenqi Fanghon recipe versus no intervention

Blinding (performance bias and detection bias)
outcome assessment

High risk

Not possible due to no intervention group and subjective assessment of xerostomia

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported (quote: "no adverse events")

Other bias

Low risk

No other sources of bias are apparent

Jaguar 2015

Methods

Location: Brazil

Number of centres: 1

Date of conduct: January 2010 to March 2012

Participants

Inclusion criteria: primary oral, oropharynx, or nasopharynx carcinomas (clinical stage ≥ II) scheduled to undergo 3‐D radiotherapy (RTC3D) or IMRT, ≤ 75 years of age

Exclusion criteria: hypersensitivity to bethanechol, hypotension, hyperthyroidism, peptic ulcer disease, epilepsy, angina, parkinsonism, and patients using tricyclic antidepressants, and antihistamines

Age: bethanechol: mean 55.9 (range 21 to 75); placebo: mean 55.8 (range 28 to 75)

Gender: bethanechol: 37 M, 11 F; placebo: 39 M, 10 F

Cancer type: oral cavity, oropharynx, nasopharynx

Radiotherapy: once‐daily mega voltage (6 MV), given at 18 to 2.12 Gy per fraction, 5 days per week (duration unclear) for 7 weeks

Chemotherapy: bethanechol 73%; placebo 71% (type of CT not reported)

Number randomised: 97 (bethanechol 48, placebo 49)

Number evaluated: 84 (bethanechol 42, placebo 42)

Interventions

Bethanechol versus placebo

Both groups: 1 tablet (25 mg) taken twice a day from beginning of RT and continued until 1 month after end of treatment (median 19 weeks)

Outcomes

Xerostomia: observer‐based grade and scored according to the subjective measures of Eisbruch (grade 0 to 3) ‐ reported as grade 2 and above

Salivary flow rates: whole unstimulated and stimulated saliva flows collected over 5 min each and reported in ml/min (reported by RT‐type subgroups ‐ we combined the subgroups but numbers were not reported so we used the number randomised from table 1), also scintigraphy undertaken

Adverse effects: bethanechol toxicities using National Cancer Institute Common Terminology Criteria for Adverse Events – NCI CTCAE, v 3.0

Survival data: not reported

Other oral symptoms: not reported

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: xerostomia assessed weekly to 3 months postRT; saliva flow assessed during RT (range 30 to 35 Gy) and 2 months postRT

Funding

FAPESP (an independent public foundation) and CAPES (an organization of the Brazilian federal government under the Ministry of Education)
Conflict of interest statement does not indicate whether there is conflict or not; quote: "All authors disclose any financial and personal relationships with other people or organizations"

Trial registration

Not registered

Sample size calculation presented

Yes (reported in supplementary data online)

Notes

Supplementary data online dx.doi.org/10.1016/j.radonc.2015.03.017

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Using the Epi‐Info® software version 6.04b, eight lists with a randomized sequence for patient allocation were generated, because a separate list was needed for each of the 8 strata defined by the 3 dichotomous stratification factors (randomization codes with block‐size of eight)"

Allocation concealment (selection bias)

Unclear risk

Authors do not state who randomised the participants and whether it was in a concealed manner

Blinding (performance bias and detection bias)
patients/carers

Low risk

Quote: "a placebo was manipulated identical in color, shape and weight. Both bethanechol and placebo therapies were coded as A and B. The clinician, patients as well as the statistician were unaware of the trial groups"

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Placebo trial

Incomplete outcome data (attrition bias)
All outcomes

Low risk

13 out of 97 dropped out with reasons for dropouts clearly stated by study group, but equal per group and similar reasons

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Jellema 2006

Methods

Location: the Netherlands

Number of centres: 1

Date of recruitment: August 1999 to August 2003

Participants

Inclusion criteria: stage III/IVB squamous cell carcinoma of the oral cavity, oropharynx, hypopharynx and/or larynx or lymph node metastases in the head and neck area from an unknown primary. Treatment with bilateral primary or postoperative radiotherapy with curative intent. 75% of the parotid gland volume expected to receive a radiation dose of at least 40 Gy. Minimal life expectancy of 12 months and a WHO performance score from 0 to 2. Good understanding of the Dutch language

Exclusion criteria: distant metastases (M1), previously irradiated patients, patients treated in combination with induction or concurrent chemotherapy, and patients with tumours that originated in the salivary glands. Pregnant patients, those participating in another investigational trial or in poor general health or psychological conditions. Patients who had severe cardiovascular disease, poor renal function or sustained hypotension not secondary to antihypertensive medication

Age: mean age = 55 (24 to 73)

Gender: AMI‐3: 20 M, 10 F; AMI‐5: 22 M, 8 F; control: 18 M, 13 F

Cancer type: head and neck at various sites and stages and lymph node classifications
Submandibular gland removal: participants were stratified by this factor but numbers of participants affected are not reported

Radiotherapy: megavolt equipment using isocentre techniques after 3‐dimensional planning. 2 opposing lateral fields with an anterior field to cover the lower jugular and supraclavicular lymph node areas. All received 46 Gy to treated areas, boost doses varied from 56 Gy (in patients who had negative surgical margins) to 63.5 Gy (in patients who had lymph node metastasis with extranodal spread or positive margins). Patients treated primarily with radiotherapy received 70 Gy to macroscopic tumour

Chemotherapy: none

Number randomised: 91 (AMI‐3: 30; AMI‐5: 30; control: 31)

Number evaluated: 71 (xerostomia at 12 months) (AMI‐3: 22; AMI‐5: 27; control: 22)

Interventions

3 arms: Amifostine 1 versus amifostine 2 versus no intervention

Group 1: amifostine 3 times weekly 200 mg/m² administered IV over 3 to 5 minutes 15 to 30 minutes before irradiation

Group 2: amifostine 5 times weekly 200 mg/m² administered IV over 3 to 5 minutes 15 to 30 minutes before irradiation
Control: nothing

Outcomes

Xerostomia: late and acute radiation‐induced xerostomia at grade 2 and above (0 to 4 scale ‐ RTOG/EORTC Late Radiation Morbidity Scoring); patient‐rated xerostomia and sticky saliva using QLQ‐H&N35 (1 to 4 scale converted linearly to a 0 to 100 mm scale where higher scores = worse symptoms) ‐ not used

Salivary flow rates: not reported

Adverse effects: vomiting (emesis), nausea, hypotension, allergic reaction

Survival data: locoregional tumour control and overall survival

Other oral symptoms: not reported

Other oral signs: not reported

Quality of life: QoL‐C30 version 3.0, the EORCT Core Questionnaire with supplemental head and neck specific module (QLQ‐H&N35)

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: xerostomia and QoL assessed at end of RT and 6, 12, 18 and 24 months after RT; survival assessed to 60 months but reported in text at 2 years

Funding

Source of funding: not stated. Amifostine provided by Schering Plough

Trial registration

Not registered

Sample size calculation presented

Reported

Notes

Have only reported locoregional tumour control and overall survival data narratively as it did not seem sensible to combine the 2 amifostine arms due to differing results. Numbers per group unclear for xerostomia at end of RT and therefore not able to use

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random assignment performed at university medical centre using a permuted block design

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding (performance bias and detection bias)
patients/carers

High risk

Blinding not possible

Blinding (performance bias and detection bias)
outcome assessment

High risk

Blinding not mentioned. Xerosomia is subjective measure

Incomplete outcome data (attrition bias)
All outcomes

High risk

22% dropouts at 12 months and difference in attrition between groups (i.e. no amifostine = 29%, amifostine3 = 27%, amifostine5 = 10%)

Selective reporting (reporting bias)

High risk

Quality of life was measured but not reported (only quote: "No significant differences")

Other bias

Low risk

No other sources of bias are apparent

Jham 2007

Methods

Location: Brazil

Number of centres: 1

Date of enrolment: October 2004 to July 2005

Participants

Inclusion criteria: adults with biopsy‐proven malignant neoplasm of the head and neck who received external beam RT

Exclusion criteria: conditions which may introduce adverse reaction to bethanechol: tricyclic antidepressants, antihistamines, betablockers, hypersensitivity

Age: bethanechol: mean 57 (SD 15); control: 55 (SD 13)

Gender: bethanechol: 17 M 5 F; control: 16 M 5 F

Cancer type: malignant neoplasm of head and neck

Radiotherapy: external beam RT, encompassing 1 or more salivary glands, minimum 45 Gy

Chemotherapy: bethanechol 23%; control 48% (type of CT not reported)

Number randomised: 43 (bethanechol 22; control 21)

Number evaluated: range over outcomes (and time points). Xerostomia VAS at 08 to 40 weeks after RT: 30 (bethanechol 13; control 17)

Interventions

Bethanechol versus artificial saliva

Bethanechol: 25 mg 3 times daily (6 am, 2 pm, 10 pm) administered with RT and used until end of RT

Control: artificial saliva (OralBalance) ‐ schedule not reported

Outcomes

Xerostomia: subjective VAS scale (length not mentioned ‐ not used), asking about dry mouth (yes/no)

Salivary flow rates: whole resting saliva and whole stimulated saliva collected over 5 minutes and reported in ml/min

Adverse effects: lacrimation, nervousness, frequent urination, sweating, warm face, cramps, diarrhoea, nausea

Survival data: death

Other oral: not reported

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: xerostomia and saliva flow assessed during RT (between 15th and 19th session), at end of RT and at least 2 months after RT (ranging from 8 to 40 weeks after)

Funding

CAPES (an organization of the Brazilian federal government under the Ministry of Education) gave financial support, Apsel Laboratories provided bethanechol, and Laclede provided artificial saliva

Trial registration

Not registered

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Using the Epi‐info software version 6.04b, 6 lists with randomized sequence for patient allocation were generated (random codes with block‐size of 8). Prior to allocation patients were stratified by RT treatment and age"

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding (performance bias and detection bias)
patients/carers

High risk

Quote: "...for obvious reasons it was not possible for the study to be double‐blinded"

Blinding (performance bias and detection bias)
outcome assessment

High risk

Quote: "...for obvious reasons it was not possible for the study to be double‐blinded"

Incomplete outcome data (attrition bias)
All outcomes

High risk

Varies over outcomes. For xerostomia (VAS): 30% dropped out or died (bethanechol 41%; control 19%). Differential dropout and (apart from death) reasons for dropouts unclear

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Lajtman 2000

Methods

Location: Croatia

Number of centres: unclear

Date of conduct: unclear

Participants

Inclusion criteria: patients scheduled to receive external beam radiation therapy to the major salivary glands completely or partially included in the field

Exclusion criteria: significant cardiovascular, pulmonary, hepatic or pancreatic disorders or gastroduodenal ulcers

Age: not reported

Gender: not reported

Cancer type: not reported

Radiotherapy: weekly external beam radiation therapy for 4 to 8 weeks, no further details

Chemotherapy: not stated

Number randomised: unclear

Number evaluated: 48

Interventions

Pilocarpine versus placebo

Pilocarpine: 5 mg capsules 4 times daily starting the day before RT and continuing for 3 months
Placebo: 5 mg capsules 4 times daily starting the day before RT and continuing for 3 months

Outcomes

Xerostomia: standardised questionnaire (subjective assessment, administered by clinician)

Salivary flow rates: stimulated salivary flow rate (parotid saliva by Carlson‐Crittenden cup; submandibular/sublingual saliva by standardised suction device)

Adverse effects: not reported

Survival data: not reported

Other oral symptoms: not reported

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: 3 months (end of drug treatment), 6 months and 12 months

Funding

Unclear

Trial registration

Unclear

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding (performance bias and detection bias)
patients/carers

Low risk

Double‐blind; pilocarpine versus placebo

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Double‐blind; pilocarpine versus placebo

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear of number randomised to each group, therefore number of dropouts unclear

Selective reporting (reporting bias)

High risk

Adverse effects not fully reported

Other bias

Low risk

No other sources of bias are apparent

Lanzós 2010

Methods

Location: Spain

Number of centres: 1

Date of enrolment: May 2004 to May 2007

Participants

Inclusion criteria: between 18 and 75 years of age. At least 10 teeth present in mouth. Willing to consent

Exclusion criteria: presence of mucosal pathology, pregnant or undergoing orthodontic therapy

Age: mouthwash: mean age 49.4 years (SD 15.4); control: mean age 54.3 years (SD 16.1)

Gender (M:F): mouthwash 15:3, control 17:1

Cancer type: head and neck

Radiotherapy: 50 to 80 Gy over 5 weeks

Chemotherapy: probably none

Number randomised: 36 (18 per group)

Number evaluated: 16 at 4 weeks for stimulated saliva (mouthwash 9, control 7)

Interventions

Antiseptic mouthrinse versus placebo

Mouthwash: CHX 0.12% and 0.05% cetylpyridinium by oral rinse 15 ml twice daily (morning and night). From start of RT for 28 days

Placebo: control without active ingredient

Outcomes

Xerostomia: not assessed

Salivary flow rates: stimulated saliva (ml/min), pH saliva (0/1/2)

Adverse effects: none reported

Survival data: not reported

Other oral symptoms: hiposialosis (drooling), mucositis, plaque, gingivitis, caries

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: 14, 28 days after RT started (i.e. no time points of interest)

Funding

Source of funding unclear; suspect pharmaceutical industry sponsored by intervention manufacture Perio‐Aid Tratamiento

Trial registration

Unclear

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer generated list assigned by random number

Allocation concealment (selection bias)

Low risk

Allocated after inclusion corresponding to numerically coded mouthrinse. Code only broken at end of study

Blinding (performance bias and detection bias)
patients/carers

Low risk

List and numbered bottles provided by promoter. Participants and researchers blinded

Blinding (performance bias and detection bias)
outcome assessment

Low risk

1 single assessor blinded to allocation

Incomplete outcome data (attrition bias)
All outcomes

High risk

56% attrition (antiseptic 50%, placebo 61%) for outcome of interest (salivary flow rates) at 4 weeks

Selective reporting (reporting bias)

High risk

Xerostomia not reported

Other bias

Low risk

No other sources of bias are apparent

Le 2011

Methods

Location: North America and Europe

Number of centres: 46 hospitals

Date of conduct: August 2005 to September 2007

Participants

Inclusion criteria: newly diagnosed unresected stage III to IV SCC of oral cavity, oropharynx, nasopharynx, hypopharynx or larynx, planned RT dose of more than 50 Gy to 2 subsites or oral cavity and oropharynx

Exclusion criteria: evidence of secondary malignancy

Age: mean 55.5 (SD 8.5)

Gender: 159 M, 29 F

Cancer type: SCC of oral cavity, oropharynx, nasopharynx, hypopharynx or larynx

Radiotherapy: mean 68 Gy in both arms for 43 days

Chemotherapy: cisplatin 100 mg/m² IV infusion on days 1, 22, and 43 of RT

Number randomised: 188 (94 per group)

Number evaluated: 188, 185 adverse events

Interventions

Palifermin versus placebo

Palifermin administered IV at 180 µg/Kg over a period of 30 to 60 seconds, in 8 weekly doses 3 days. Bolus injection before radiotherapy, then at weekend

Placebo: matching as above (1.2 ml of sterile water +)

Follow‐up: median follow‐up 25.9 months palifermin, 25.0 placebo

Outcomes

Xerostomia: incidence of grade ≥ 2 xerostomia (Common Terminology Criteria for Adverse Events (CTCAE) v 3.0 Dry Mouth/Xerostomia scale ‐ info from trials registry), assessed at months 4, 6, 8, 10, 12, reported only at month 4

Salivary flow rates: not reported

Adverse effects: assessed by Common Terminology Criteria for Adverse Events: nausea, constipation, decreased weight, vomiting, anaemia, leukopenia, fatigue, dehydration

Survival data: overall tumour response, time to locoregional tumour failure, incidence of secondary primary tumours, overall and progression‐free survival

Other oral symptoms: dysphagia (difficulty in swallowing), OMWQ‐HN 0 (no soreness) to 4 (extreme soreness) scale for mouth and throat soreness (assessed twice weekly by trained evaluators during radiochemotherapy)

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Funding

Externally funded by Amgen GSK

Trial registration

NCT00101582 (clinicaltrials.gov/show/NCT00101582)

Sample size calculation presented

Reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Centralised randomisation system. 1:1 allocation ratio

Allocation concealment (selection bias)

Low risk

Centralised randomisation for all sites, probably allocation concealment

Blinding (performance bias and detection bias)
patients/carers

Low risk

Quote: "placebo‐controlled, double‐blind study"
Comment: blinding feasible

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Quote: "placebo‐controlled, double‐blind study"
Comment: blinding feasible

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All cases accounted for. ITT analysis (patients having no assessment assumed to have event)

Selective reporting (reporting bias)

Low risk

All outcomes reported. Low risk for xerostomia at the time point used in meta‐analysis (4 months) ‐ however, it should be noted that xerostomia was measured up to 12 months but data not reported

Other bias

Low risk

No other sources of bias are apparent

Lin 2014

Methods

Location: Taiwan, Republic of China

Number of centres: 1

Date of enrolment: January 2003 to November 2004

Participants

Inclusion criteria: histological evidence of carcinoma of head and neck, to receive RT, life expectancy ≥ 3 months, ECOG status ≤ 2. Criteria such as white blood cells, platelets, haemoglobin had to be within certain parameters

Exclusion criteria: prior RT, presence of oral lesions, severe organ failure, brain metastasis

Age: TWBXM: mean 51 (SD 15); placebo: 54 (SD 16)

Gender: TWBXM: 29 M, 9 F; placebo: 32 M 3 F

Cancer type: treatment group: head and neck cancer stage 0 = 4, I = 4, II = 10, III = 4, IVA = 12, IVB = 4. Control group: head and neck cancer stage 0 = 3, I = 5, II = 8, III = 4, IVA = 10, IVB = 5

Radiotherapy: TWBXM: mean dose 6944.9 cGy; placebo: mean dose 7098.4 cGy

Chemotherapy: not mentioned probably not given

Number randomised: 73 (TWBXM 38; placebo 35)

Number evaluated: 71 (TWBXM 38; placebo 33)

Interventions

Traditional Chinese medicine (TWBXM) versus placebo

Tianwang Buxin Mini‐pills (TWBXM) ‐ 13 herbs listed in paper

Placebo made of starch and designed to taste and look similar to intervention

3 g orally 3 times daily starting from initiation of RT until 1 month after RT completion

Follow‐up: end of RT (end of RT for dichotomous data, 1 month postRT for continuous data)

Outcomes

Xerostomia: RTOG grades by evaluating physician ‐ none, slight, moderate mouth dryness

Salivary flow rates: not reported

Adverse effects: EORTC QLQ‐C30 ‐ skin, nausea, vomiting, leukopenia, fatigue, pain, dyspnoea, insomnia, appetite loss, constipation, diarrhoea, body weight loss

Survival data: not reported

Other oral symptoms: EORTC QLQ‐C30 ‐ pain, swallowing, speech problems, mouth opening, teeth, mucositis, oral mucosa, loss of taste

Other oral signs: not reported

Quality of life: EORTC QLQ‐C30 and head and neck specific QLQ‐H & N35

Patient satisfaction: not reported

Cost data: not reported

Funding

Government. Committee on Chinese Medicine and Pharmacy, Department of Health, Executive Yuan, Taiwan (grants CCMP92‐RD‐011 and CCMP93‐RD‐008). Quote: "All authors declare that there are no conflicts of interest"

Trial registration

Not registered

Sample size calculation presented

No

Notes

Problematic data ‐ emailed corresponding author 22 February 2016

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "...randomized to study medicine according to a computer‐generated randomization schedule..."

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding (performance bias and detection bias)
patients/carers

Low risk

Quote: "All patients, the study nurses and doctors were blinded to the group of the treatment group"

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Quote: "All patients, the study nurses and doctors were blinded to the group of the treatment group"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

The dropout rate was 3% to 7% (data reported inconsistently in the report) at the end of RT, which is the preferred time point for this review. However, there were 29 (40%) dropouts at the time point of 1 month postRT

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Lozada‐Nur 1998

Methods

Location: USA

Number of centres: 1

Date of conduct: not stated

Participants

Inclusion criteria: not reported

Exclusion criteria: not reported

Age: pilocarpine: mean 51.5 years (range 29 to 76); placebo: 54.8 years (range 47 to 68)

Gender (M:F): pilocarpine 9:2, placebo 9:2

Cancer type: nasopharyngeal cancer

Radiotherapy: total dose 60 to 70 Gy

Chemotherapy: some

Number randomised: 22 (11 per group)

Number evaluated: 11 per group for incidence of dry mouth. There are discrepancies over the numbers

Interventions

Pilocarpine versus placebo

Pilocarpine: 5 mg tablets 3 times daily ‐ 4 times daily, 2 weeks before RT and concurrently with RT

Placebo

Outcomes

Xerostomia: questionnaire

Salivary flow rates: resting salivary flow

Adverse effects: sweating, lacrimation (excess tears, crying), rhinorrhoea (watery discharge from the nose), diarrhoea, nausea, rhinitis, blurred vision, constipation, neuropathy

Survival data: not reported

Other oral symptoms: mucositis, dysphagia (difficulty in swallowing), dysgeusia (taste disturbance), pain

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: prior to radiation, weekly during treatment, end of RT, 3 months after RT

Funding

Supported by a grant from MGI Pharma Inc.

Trial registration

Not registered or published

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Contact with authors confirmed that the random allocation was conducted by pharmaceutical company, producing coded containers

Allocation concealment (selection bias)

Low risk

Quote: "Allocation sequence was provided in a logo‐type format and kept by our dental assistant (in a locked cabinet)"
Comment: further description of this from study authors implies allocation concealment

Blinding (performance bias and detection bias)
patients/carers

Low risk

Quote: "double‐blind"

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Quote: "double‐blind"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

2 dropped out because of adverse events (severe nausea and vomiting, and tumour growth). Unclear reasons for all dropouts for xerostomia outcome (6)

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Patni 2004

Methods

Location: India

Number of centres: 1

Date of conduct: not reported

Participants

Inclusion criteria: locally advanced histologically proven squamous cell carcinoma of head and neck region. 75% or more of each parotid gland was included in the radiation portal. Age above 18 years. Expected survival > 12 months. Karnofsky Performance Status > 60. Normal haemogram, renal and liver functions, normal calcium levels

Exclusion criteria: prior treatment for malignancy, associated hypotension or distant metastases

Age: not reported

Gender: 65% M

Cancer type: head and neck

Radiotherapy: external radiation therapy with gamma rays to a dose of 66 to 72 Gy with conventional fractionation

Chemotherapy: 40 mg/m² cisplatin weekly

Number randomised: 170 (85 per group)

Number evaluated: 170 (85 per group)

Interventions

Amifostine versus no intervention

Amifostine: 250 mg IV over 3 minutes for 4 days a week from day 1 of radiotherapy until completion of treatment
Control: nothing

Outcomes

Xerostomia: acute and late xerostomia grade 2 and above (RTOG 0‐4 scale)

Salivary flow rates: parotid scintigraphy (no data)

Adverse effects: not reported

Survival data: disease‐free survival at 24 months and tumour response

Other oral symptoms: mucositis (RTOG)

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported
Timing of assessments: xerostomia at 3 and 12 months after RT; parotid scintigraphy at 3, 6, 12 and 24 months after RT; survival at 24 months

Funding

Not reported

Trial registration

Not registered or published

Sample size calculation presented

No

Notes

Abstract with additional information provided by study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised but unclear method

Allocation concealment (selection bias)

Unclear risk

Not mentioned

Blinding (performance bias and detection bias)
patients/carers

High risk

Not possible due to 'no treatment' group

Blinding (performance bias and detection bias)
outcome assessment

High risk

Not possible due to 'no treatment' group. Xerostomia is subjective outcome

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

High risk

No information on adverse events

Other bias

Low risk

No other sources of bias are apparent

Peng 2006

Methods

Location: China

Number of centres: 1

Date of conduct: October 2003 to October 2005

Participants

Inclusion criteria: quote: "1) H&N SC patients diagnosed with pathological examinations, who cannot endure surgery or cannot be treated with radical resection; 2) first‐visit patients who have not received cancer treatment, with no tumour metastasis; 3) WBC > 4.0*109/L, platelet count > 100.0*109/L, Hb > 10 g/L, normal function of heart, lungs, liver and kidneys; 4) Karnofsky score ≥ 60; 5) patients gave informed consent"

Exclusion criteria: see above

Age at baseline (years): amifostine: median 58; control: median 57

Gender: amifostine: 12 M, 6 F; control: 12 M, 6 F

Cancer type: head and neck squamous carcinoma (amifostine/control): oral = 4/3, nasopharyngeal = 6/6, oropharyngeal = 5/5, hypopharyngeal = 1/1, laryngeal = 1/1, paranasalsinus = 1/1, glottis = 0/1

Radiotherapy: quote: "Primary site of tumour and cervical lymph nodes: 74.4 Gy overall, 1.2 Gy per time, 2 times per day (> 6 hours between these 2 times), for 4 to 5 weeks; separate routine fractionated RT for the neck area: 50 Gy overall, 25 times, 5 weeks"

Chemotherapy: quote: "Continued IV infusion of 5 FU 750 mg/m² using pump, 24 hours per day for 3 days. On the 5th day after 5 FU use, intravenous infusion of cisplatin 50 mg/m² with 250 or 500 ml saline (2 to 4 hours). IV infusion of docetaxel 75 mg/m² with 250 ml saline (< 1.5 hours). Chemotherapy performed in rounds: 1st round – during RT, 2nd round – begin in the 5th week after RT, 3rd round – begin in the 9th week after RT"

Number randomised: 37 (amifostine 18, control 19)

Number evaluated: 36 (amifostine 18, control 18)

Interventions

Amifostine versus no intervention

Amifostine: quote: "400 mg amifostine each time, intravenous infusion 15 minutes before RT and chemotherapy (finish in 5 to 7 minutes)"

Control: no intervention other than the same RT and chemotherapy

Outcomes

Xerostomia: only results for 'acute' and 'chronic' dry mouth reported; time and standards for assessments not described

Salivary flow rates: not reported

Adverse effects: hypotension, nausea, vomiting, dizziness, fatigue, hiccup, sneezing, facial flush

Survival data: not reported

Other oral symptoms: mucositis

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: not reported (therefore unable to use data)

Funding

Not reported; conflicts of interest: not reported

Trial registration

Not registered

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "patients were randomized to"

Allocation concealment (selection bias)

Unclear risk

No details given

Blinding (performance bias and detection bias)
patients/carers

High risk

Amifostine versus no intervention. Blinding not possible

Blinding (performance bias and detection bias)
outcome assessment

High risk

Blinding not mentioned. Xerostomia is subjective measure

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Quote: "1 patient in the amifostine group quitted due to financial reasons"

Selective reporting (reporting bias)

Unclear risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Pimentel 2014

Methods

Location: Brazil

Number of centres: 1

Date of conduct: not reported

Participants

Inclusion criteria: newly diagnosed head and neck cancer beginning treatment with RT

Exclusion criteria: previous RT, concomitant chemotherapy, cardiopathy, hypertension, diabetes, allergy to pilocarpine, Sjögren syndrome, salivary gland tumours, chronic lung disease, glaucoma, peptic ulcer, taking betablockers or drugs that could alter salivary flow

Age: mean 60 years (not given by group)

Gender (M:F): 8:3

Cancer type: oral (n = 1); oropharynx (n = 3); mouth floor and tongue (n = 2); larynx (n = 4); pharynx (n = 1)

Radiotherapy: 35 to 50 Gy, with daily doses about 2 Gy

Chemotherapy: none

Number randomised: unclear whether 29 or 11 (see attrition bias)

Number evaluated: 11 (pilocarpine 5, placebo 6)

Interventions

Pilocarpine versus placebo

Pilocarpine: 5 mg 3 times daily for duration of RT

Placebo: saline solution 3 times daily for duration of RT

Outcomes

Xerostomia: patient‐reported feeling of dry mouth

Salivary flow rates: unstimulated (USF) and stimulated saliva (SSF) (ml/min)

Adverse effects: reported narratively

Survival data: locoregional control: not reported

Other oral symptoms: oral mucositis, ulcers

Other oral signs: difficulty in eating

Quality of life: only eating

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: weekly during RT (4 weeks)

Funding

The National Research Council (CNPq)

Trial registration

Not registered

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "...dispensing pharmacy held custody of the samples, separating those from the group who took pilocarpine solution from those which took the placebo. All patients were assigned a number corresponding to the medicine bottle. Researchers were not granted access to that information prior to the end of the survey"

Allocation concealment (selection bias)

Low risk

Quote: "...dispensing pharmacy held custody of the samples, separating those from the group who took pilocarpine solution from those which took the placebo. All patients were assigned a number corresponding to the medicine bottle. Researchers were not granted access to that information prior to the end of the survey"

Blinding (performance bias and detection bias)
patients/carers

Low risk

Double‐blind ‐ see above

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Double‐blind ‐ see above

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "We pre‐selected 29 patients; however, the careful selection of the population was directly reflected in the number of enrolled patients and in the end, only 11 were included in the survey. We consider that this low number is a result not only of the exclusion stemming from previously established eligibility criteria but also from the breach of protocol"

Selective reporting (reporting bias)

High risk

Poorly reported data for xerostomia and no adverse events

Other bias

Low risk

No other sources of bias are apparent

Reshma 2012

Methods

Location: India

Number of centres: 1

Date of conduct: not stated

Participants

Inclusion criteria: carcinoma of the head and neck stage III and IV, aged between 30 and 70, to receive RT, normal haematology, biochemistry and Karnofsky Performance Index > 70%

Exclusion criteria: poor general condition, associated co‐morbidities, psychiatric conditions

Age: 30‐70

Gender: unclear

Cancer type: SCC of head and neck

Radiotherapy: 60 Gy on cobolt 60 for 30 days over 6 weeks

Chemotherapy: not reported but probably none

Number randomised: 20 or 40 unclear

Number evaluated: no apparent dropouts but unclear how many started

Interventions

Tulasi (Ocimum Sanctum) versus placebo

Tulasi (Ocimum Sanctum)

Placebo: vitamin B complex

2 capsules of 250 mg orally half an hour prior to RT

*Healthy control group also included but not used (not randomised)

Follow‐up: 29 days of RT

Outcomes

Xerostomia: unclear, quote: "patients were assessed at the end of every week for grade of mucositis, skin reaction and salivary status"

Salivary flow rates: not reported

Adverse effects: not reported

Survival data: not reported

Other oral symptoms: not reported

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Funding

Not reported

Trial registration

Not registered

Sample size calculation presented

No

Notes

No useable data ‐ emailed corresponding author 27 January 2016

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were randomized into 2 arms"
Comment: insufficient information on method of random sequence generation

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding (performance bias and detection bias)
patients/carers

Low risk

Placebo‐controlled

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Placebo‐controlled

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

High risk

No adverse event data reported. Poorly reported xerostomia

Other bias

Low risk

No other sources of bias are apparent

Rode 1999

Methods

Location: Slovenia

Number of centres: 1

Dates and duration of recruitment period: not stated

Participants

Inclusion criteria: irradiated for head and neck cancer and salivary glands included in the irradiation field

Exclusion criteria: not reported

Age: aged 32 to 72, median 62 years

Gender (M:F): 60:9

Cancer type: oral cavity (n = 14), oropharynx (n = 33), hypopharynx (n = 8), larynx (n = 11), other (n = 3) (evenly distributed across groups)

Radiotherapy: 5 Gy per day, 5 days a week. Irradiated volume reduced twice during irradiation treatment: at 40 Gy for shielding the spinal cord and at 60 Gy for treating the area of original disease, up to 70 Gy. Postoperative patients received 50 Gy ‐ 56 Gy with parallel opposed portals only. 44 patients had postoperative RT and 25 were treated by RT alone
Mean irradiation dose (Gy) delivered to area of salivary glands

Chemotherapy: not stated

Number randomised: 69 (A: 9, B: 30, C: 30)

Number evaluated: 69 (A: 9, B: 30, C: 30)

Interventions

Pilocarpine (postRT) + Biperiden (during RT) versus no intervention

Group Aª: pilocarpine during RT and 6 weeks after. Pilocarpine hydrochloride (5 mg) perorally 3 times daily administered 1 hour before irradiation
Group B: Biperiden during RT and pilocarpine after RT group. Biperidin chloride (2 mg tablets) 1 and a half hours before irradiation, and pilocarpine hydrochloride (5 mg 3 times daily) for 6 weeks after RT
Group C: no intervention

Outcomes

Xerostomia: not reported

Salivary flow rates: mean quantity of non‐stimulated saliva secretion (ml/min)

Adverse effects: not reported

Survival data: not reported

Other oral symptoms: mucositis, swallowing WHO criteria

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: end of RT, 3 months, 6 months and 12 months after end of RT

Funding

Source of funding: none

Trial registration

Not registered

Sample size calculation presented

No

Notes

ªGroup A data not used: randomisation to Group A was stopped after the first 9 patients for ethical reasons ‐ 3 months after RT total cessation of saliva secretion was observed in all except 1 patient

Follow‐up: 12 months

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Information provided by author: "Sequence centrally generated"

Allocation concealment (selection bias)

Low risk

Author included the following in an email "randomization with permuted blocs, participants were allocated to the treatment groups randomly. Allocation sequence was generated centrally, treating physician (radiologist) enrolled patient and participants were assigned to the groups by specialist in dental medicine"

Comment: centralised random allocation and was probably adequately concealed

Blinding (performance bias and detection bias)
patients/carers

High risk

No blinding undertaken

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Salivary flow objective measure

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised patients included in analysis

Selective reporting (reporting bias)

High risk

Xerostomia and adverse events not reported

Other bias

Low risk

No other sources of bias are apparent

Sangthawan 2001

Methods

Location: Thailand

Number of centres: 1

Date of recruitment: January 1998 to January 1999

Participants

Inclusion criteria: histologically documented squamous cell carcinoma of head and neck who would receive definite or postoperative radiation

Exclusion criteria: significant uncontrolled cardiac, pulmonary, renal or occular disease or required tricyclic antidepressants or antihistamine with anticholinergic effects, betablocker, pilocarpine for ophthalmic indications or chemotherapy

Age: pilocarpine: 57 years; placebo: 58 years

Gender (M:F): 49:11

Cancer type: oropharynx (n = 27); nasopharynx (n = 14); others (n = 19)

Radiotherapy: Cobalt‐60 or 6 MV photon machine. Standard arrangement ‐ opposing lateral portals, loaded 1:1 and/or anterior low neck field. Both parotids treated to a dose of at least 50 Gy with an equal daily dose of 1.8‐2.0 Gy

Chemotherapy: none

Number randomised: 60 (30 per group)

Number evaluated: 47 (25 pilocarpine; 22 placebo)

Interventions

Pilocarpine versus placebo

Pilocarpine jelly: self administered 5 mg 3 times daily at meal times for duration of RT (7 weeks)
Placebo: self administered 3 times daily at meal times for duration of RT (7 weeks)

Follow‐up: 6 months after RT

Outcomes

Xerostomia: subjective evaluation scores for xerostomia questionnaire (100 mm VAS for each of 5 questions)

Salivary flow rates: not reported

Adverse effects: reported ("non‐specific symptoms such as nausea, vomiting, dizziness, urinary frequency, palpitation, sweating

and tearing")

Survival data: not reported

Other oral symptoms: not reported

Other oral signs: disability to oral intake, amount of meals, use of analgesics

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: 6 months after RT

Funding

Faculty of Medicine, Prince of Songkla University

Trial registration

Not registered

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Information provided by author: "random number table"

Allocation concealment (selection bias)

Low risk

Information provided by author: "clinician not participating in study generated allocation sequence. Treatment codes concealed in sealed envelopes. Treatment coding not disclosed to investigator or patient"

Blinding (performance bias and detection bias)
patients/carers

Low risk

Quote: "identically appearing placebo... patients and investigators were unaware of which treatment was administered"

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Information provided by author: "treatment code was disclosed to the investigator only after completion of the analysis of the results of the study"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

22% dropout rate

Selective reporting (reporting bias)

High risk

Poor reporting of xerostomia without SD

Other bias

Low risk

No other sources of bias are apparent

Vacha 2003

Methods

Location: Germany

Number of centres: 1

Date of conduct: October 1996 to February 1999

Participants

Inclusion criteria: advanced tumours of larynx, oro or hypopharynx, all received surgery and were to receive RT plus CT, pathologically confirmed cancer, between 18 and 70 years, performance status WHO 0‐II, adequate bone marrow and liver and renal function

Exclusion criteria: severe internal medical disorders, hyper or hypotension, history of RT or CT, women with inadequate contraception, secondary malignancies, recurrent tumours

Age: amifostine: mean 53.5; control: mean 55.1

Gender: amifostine: M 21, F 4; control: M 19, F 6

Cancer type: amifostine: SSC (24) and adenocarcinoma (1); control: SSC (24) and lymphoepithelial cancer (1)
Neck dissection involving removal of submandibular glands: radical bilateral (amifostine 32%, control 40%); radical unilateral + selective contralateral (amifostine 16%, control 12%); radical unilateral (amifostine 36%, control 16%)

Radiotherapy: conventionally fractionated RT (5 x 2 Gy/week). Total dose 60 Gy for completely resected tumours, 70 Gy incomplete resected tumours

Chemotherapy: individually planned. 70 mg/m² carboplatin on treatment days 1 to 5 and 29 to 33 just before RT session (all participants had this)

Number randomised: 56 (not reported by group)

Number evaluated: 50 (25 per group); 41 (amifostine 19, control 22) for xerostomia

Interventions

Amifostine versus no intervention

250 mg amifostine given intravenously as short infusion over 10 to 15 minutes

Outcomes

Xerostomia: RTOG (not useable)

Salivary flow rates: not reported

Adverse effects: skin toxicity, loss of hair

Survival data: not reported

Other oral symptoms: oral mucositis

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: weekly during RT until week 6 (during RT; no usable data)

Funding

Not reported

Trial registration

Not registered

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "...were randomized to receive..."

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding (performance bias and detection bias)
patients/carers

High risk

No treatment control group

Blinding (performance bias and detection bias)
outcome assessment

High risk

Xerostomia is a subjective assessment by the patient

Incomplete outcome data (attrition bias)
All outcomes

High risk

Reasons for dropouts by study group unclear. 6 dropouts altogether: allergic skin reaction (1) after 5th application of amifostine, patient refusal (3), second malignancy (1), surgical complications (1). 27% attrition overall for xerostomia ‐ participants with larynx cancer excluded from analysis

Selective reporting (reporting bias)

High risk

Xerostomia selectively and poorly reported (only significant data reported)

Other bias

Low risk

No other sources of bias are apparent

Valdez 1993

Methods

Location: USA

Number of centres: multisite unclear how many

Date of conduct and duration of recruitment period: not stated

Participants

Inclusion criteria: scheduled to receive external beam radiation therapy to the major salivary glands, completely or partially

Exclusion criteria: significant cardiovascular, pulmonary, hepatic, or pancreatic disorders or gastroduodenal ulcers. Women with childbearing potential were required to have a pregnancy test with negative results before entry and to use contraception during the study

Age: pilocarpine: 22 to 65 years, mean 42.6 years; placebo: 21 to 56 years, mean 40.2 years

Gender (M:F): 6:4

Cancer type: mix of SCC, mucoepidermoid carcinoma, Hodgkin disease and malignant lymphoma

Radiotherapy: mean dose 41.9 Gy

Chemotherapy: not stated

Number randomised: 10 (5 per group)

Number evaluated: 9 (pilocarpine 5; placebo 4)

Interventions

Pilocarpine versus placebo

Pilocarpine: 5 mg capsules 4 times daily for 3 months starting the day before RT
Placebo: 5 mg capsules 4 times daily for 3 months starting the day before RT
All participants received a rigorous preventative oral hygiene regimen including topical fluoride applications

Outcomes

Xerostomia: subjective assessment using questionnaire

Salivary flow rates: stimulated salivary flow rates (μl/min)

Adverse effects: not reported

Survival data: quote: "all tumour responded favourably and all were in complete remission for the remainder of the study"

Other oral symptoms: not reported

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: 3, 4, 5, 6 and 12 months from start of RT

Funding

National Institute of Dental and Craniofacial Research (NIDCR). Randomisation sequence generation and allocation were undertaken by US National Institutes of Health (NIH) pharmaceutical service

Trial registration

Not registered

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sequence generation not explicit but conducted by NIH Pharmaceutical Development Service

Allocation concealment (selection bias)

Low risk

Conducted by NIH Pharmaceutical Development Service; intervention dispensed in coded bottles

Blinding (performance bias and detection bias)
patients/carers

Low risk

Pilocarpine versus placebo

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Participants are blinded and providing saliva and questionnaire data

Incomplete outcome data (attrition bias)
All outcomes

Low risk

1/5 participants in placebo group dropped out. No dropouts in pilocarpine group

Selective reporting (reporting bias)

High risk

No adverse event data reported

Other bias

Low risk

No other sources of bias are apparent

Veerasarn 2006

Methods

Location: Thailand

Number of centres: 5

Date of recruitment: February 1999 to September 2001

Participants

Inclusion criteria: histological proven squamous cell carcinoma of head and neck region; ECOG performance statue 0‐2; adequate bone marrow, liver and renal functions; age 18‐70 years; no prior definite/radical surgery, chemotherapy, radiotherapy or biological response modifier; no evidence of distant metastasis; life expectancy ≥ 12 months; able to comply with a follow‐up schedule; weight loss ≤ 10% in previous 3 months

Exclusion criteria: concomitant malignant disease in other parts of the body; active uncontrolled infection; pregnant or lactating women; medical or psychiatric illness that compromise the patient's ability to complete the study; concomitant use of chemotherapy

Age: amifostine: mean 55 (23‐70); control: mean 52 (23 to 69)

Gender: amifostine: 24 M, 8 F; control: 27 M, 8 F

Cancer type: oral cavity, oropharynx, nasopharynx, larynx, hypopharynx

Radiotherapy: standard fractionation (2 Gy, 5 days a week). Duration: 5 to 8 weeks. Definite RT = 70 Gy. Postoperative RT = 50 Gy. Amifostine group: definite RT = 15; postoperative RT = 17. Control group: definite RT = 18; postoperative RT = 17
Chemotherapy: none

Number randomised: 67 (amifostine 32, control 35)

Number evaluated: 62 (amifostine 32, control 30)

Interventions

Amifostine versus no intervention

Amifostine: (Ethyol) (200 mg/m²) IV (3 to 5 min), 30 min before RT. 5 consecutive days a week for 5 to 7 weeks (during RT)
Control: nothing

Outcomes

Xerostomia: 1) questionnaire (6 questions) (RTOG/EORTC acute and late radiation morbidity scoring criteria) for xerostomia: dryness of mouth, oral comfort, quality of sleep, ability to speak, ability to chew and swallow and ability to wear dentures (average score 0 to 10: 0 = normal); 2) RTOG 0 to 4 scale ‐ grade 2 and above

Salivary flow rates: unstimulated and stimulated whole saliva collection (mg/5 min) and scintigraphy

Adverse effects: nausea, vomiting, hypotension

Survival data: disease‐free survival

Other oral symptoms: mucositis (RTOG grade 2‐3)

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessments: weekly during RT (for 6 weeks), then at end of RT and at 1, 2, 3, 6, 12, 18 and 24 months after RT; survival at 24 months after RT

Funding

Source of funding: unclear

Trial registration

Not registered or published

Sample size calculation presented

No

Notes

Information on randomisation, and numbers and SDs from correspondence

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random number table

Allocation concealment (selection bias)

Low risk

Central allocation

Blinding (performance bias and detection bias)
patients/carers

High risk

Amifostine versus no intervention

Blinding (performance bias and detection bias)
outcome assessment

High risk

Xerostomia is subjective measure

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "We excluded 5 cases in the control as they did not have salivary gland function, or had severe salivary gland impairment"

Comment: we are assuming that there were no other dropouts (assessment made on 32 in amifostine, 30 in control)

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Wang 1998

Methods

Location: China

Number of centres: 1

Date of recruitment: May 1996 to October 1997

Participants

Inclusion criteria: patients with head and neck cancer, pathology confirmed, aged 20 to 70 and radiation fields including the main salivary glands, total dose > 60 Gy

Exclusion criteria: severe systemic disease or history of chronic diseases of the salivary glands

Age (years): treatment: mean 46.8 (range 22 to 68); placebo: mean 45.5 (range 24 to 70)

Gender: treatment: 20 M, 4 F; placebo: 21 M, 5 F

Cancer type: treatment: nasopharyngeal = 17, throat = 4, tonsil = 3; placebo: nasopharyngeal = 18, throat = 4, tonsil = 3, hard palate = 1

Radiotherapy: 6 MV x‐ray with accelerator linear, SL‐75 produced by Philips. 2 Gy, 5 times per week for 6 to 7 weeks (total dose 60‐70 Gy)

Chemotherapy: none

Number randomised: 50 (treatment 24, placebo 26)

Number evaluated: 50 (treatment 24, placebo 26)

Interventions

Chinese medicine versus placebo (Dobell's solution)

Chinese medicine and Dobell's solution (20 ml 3 times daily) rinsing and spray inhalation for the duration of the RT starting from the beginning of RT
Chinese medicine: formulation of fragrant solomonseal rhizome (30 g), dwarf lilyturf tuber root (20 g), peach seed (24 g), dendrobium stem (30 g), wolfberry fruit (30 g), rehmannia dried root (40 g), prepared rehmannia root (40 g), American ginseng (30 g), safflower (20 g), chuanxiong rhizome (20 g). Mixture broken into a powder, soaked in 1000 ml of water, 40 degrees Centigrade for 6 hours, ultrasound oscillation for 10 minutes, centrifugation and the liquid part is used for clinical use

Placebo: Dobell's solution

Follow‐up: end of RT

Outcomes

Xerostomia: subjective evaluation score (VAS) for xerostomia, before and during the RT (at 10, 20, 30, 40, 50 and 60 Gy)

Salivary flow rates: stimulated salivary flow rates, in morning around 9 am (at least 1 hour after breakfast) patients rinsed with water, then chewed gum and saliva collected after 5 minutes, before and during the RT (at 20, 40 and 60 Gy)

Adverse effects: not reported

Survival data: not reported

Other oral symptoms: not reported

Other oral signs: not reported

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Funding

Not reported; conflicts of interest: not reported

Trial registration

Not registered

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomly divided"

Comment: no further details given

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding (performance bias and detection bias)
patients/carers

Low risk

Placebo used

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Placebo used

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

High risk

Adverse effects not reported. Data for xerostomia not usable (reported as a graph with no SDs)

Other bias

Low risk

No other sources of bias are apparent

Warde 2002

Methods

Location: Canada

Number of centres: 1

Date and duration of recruitment period: not stated

Participants

Inclusion criteria: squamous cell head and neck cancer, scheduled to receive RT with the inclusion of > 50% of both parotid glands in the radiation fields to doses > 50 Gy. Primary treatment and postoperative RT participants

Exclusion criteria: previous RT or chemotherapy or pre‐existing xerostomia from other causes. Medical contraindication to pilocarpine

Age: pilocarpine: mean 56.2 years; placebo: mean 57.8 years

Gender (M:F): 94:36

Cancer type: SSC of head and neck

Radiotherapy: 60‐70 Gy in 2 Gy daily fractions (68 participants), 60‐64 Gy in 40 fractions during 4 weeks using twice daily treatments (33 participants), 50 Gy in 25 daily fractions (7 participants), 60 Gy in 25 daily fractions (8 participants) and 51 Gy in 20 daily fractions (7 participants)

Chemotherapy: not stated

Number randomised: 130 (65 per group)

Number evaluated: for xerostomia: 92 (pilocarpine 48, placebo 44) at 3 months postRT; 87 (pilocarpine 46, placebo 41) at 6 months postRT

Interventions

Pilocarpine versus placebo

Pilocarpine: 5 mg tablets 3 times daily starting day 1 of RT and continued until 1 month after completion of RT
Placebo: tablets 3 times daily starting day 1 of RT and continued until 1 month after completion of RT

Outcomes

Xerostomia: VAS (patient‐completed) assessing patient's perception of dryness of their mouth (7 questions). Scores from 0 to 100, low scores = most difficulty

Salivary flow rates: not reported

Adverse effects: excessive sweating, acute toxicity of therapy (RTOG)

Survival data: not reported

Other oral symptoms: not reported

Other oral signs: feeding tube inserted

Quality of life: patients' quality of life (McMaster University Head and Neck Questionnaire (HNRQ)). Score 1‐7, lower score = poorer quality of life

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: 1, 3 and 6 months after end of RT

Funding

Unrestricted educational grant from Pharmacia Canada

Trial registration

Not registered

Sample size calculation presented

Yes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding (performance bias and detection bias)
patients/carers

Low risk

Pilocarpine versus placebo, "double‐blind"

Blinding (performance bias and detection bias)
outcome assessment

Low risk

Self reported

Incomplete outcome data (attrition bias)
All outcomes

High risk

130 participants randomised. 19/65 dropouts in pilocarpine group; 24/65 dropouts in placebo group. 8 in pilocarpine and 4 in placebo dropped out for toxicity, otherwise reasons unclear

Selective reporting (reporting bias)

Low risk

Xerostomia and adverse events reported

Other bias

Low risk

No other sources of bias are apparent

Watanabe 2010

Methods

Location: Japan

Number of centres: 1

Date of conduct: January and October 2009

Participants

Inclusion criteria: head and neck cancer scheduled for RT or RT + CT

Exclusion criteria: not reported

Age: polaprezinc: 67.4 (range 53 to 78); control: 62.7 (range 35 to 86)

Gender (M:F): polaprezinc 13:3, control 11:4

Cancer type: head and neck, mainly pharyngeal and laryngeal

Radiotherapy (mean dose and duration): polaprezinc: 51 Gy (range 30‐70), 37 days (range 21‐55); control: 58 Gy (range 36‐70), 45 days (range 28‐79)

Chemotherapy: some but unclear what, concomitantly carried out for 56% of polaprezinc and 80% of control

Number randomised: 31 (polaprezinc 16, control 15)

Number evaluated: 31 (polaprezinc 16, control 15)

Interventions

Polaprezinc versus azulene oral rinse

Polaprezinc granules (0.5 g) were dissolved in 20 ml of 5% sodium alginate

Azulene oral rinse prepared by pouring 7 drops of 4% solution into 100 ml water

Both groups administered via oral rinse. Rinse for 3 minutes, 4 times daily, from start to end RT. After rinsing, polaprezinc swallowed but azulene spat out. From start to end of RT

Outcomes

Xerostomia: CTCAE 0‐3 grade

Salivary flow rates: not reported

Adverse effects: not reported

Survival data: tumour response by RECIST criteria for specific group of patients

Other oral symptoms: pain, dysgeusia (taste disturbance), mucositis

Other oral signs: amount of meals

Quality of life: not reported

Patient satisfaction: not reported

Cost data: not reported

Timing of assessment: over RT period (maximum score)

Funding

Source of funding unclear

Trial registration

Unclear

Sample size calculation presented

No

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "...were randomly assigned". No details given

Allocation concealment (selection bias)

Unclear risk

No details given

Blinding (performance bias and detection bias)
patients/carers

High risk

None. Polaprezinc swallowed after rinsing, while azulene is spat out. Quote: "open trial"

Blinding (performance bias and detection bias)
outcome assessment

High risk

Xerostomia subjective measurement, and different interventions

Incomplete outcome data (attrition bias)
All outcomes

Low risk

None

Selective reporting (reporting bias)

High risk

No adverse events reported

Other bias

Low risk

No other sources of bias are apparent

CHX = chlorhexidine; CRT = chemoradiotherapy; CT = chemotherapy; CTCAE = Common Terminology Criteria for Adverse Events; ECOG = Eastern Cooperative Oncology Group score; EORTC = European Organisation for Research and Treatment of Cancer; F = female; G‐CSF = granulocyte‐colony stimulating factor; GI = gastrointestinal; GM‐CSF = granulocyte‐macrophage colony‐stimulating factor; Gy = gray; HNSCC = head and neck squamous cell carcinoma; IMRT = intensity‐modulated radiation therapy; ITT = intention‐to‐treat; IV = intravenous; KGFs = keratinocyte growth factors; M = male; min = minute; MV = megavolt; NCI CTC = National Cancer Institute Common Toxicity Criteria; NCIC CTG ECTC = National Cancer Institute of Canada Clinical Trials Group Expanded Common Toxicity Criteria; OMAS = Oral Mucositis Assessment Scale; QoL = quality of life; RECIST = Response Evaluation Criteria In Solid Tumours; RT = radiotherapy; RTOG = Radiation Therapy Oncology Group; SC = subcutaneous; SCC = squamous cell carcinoma; SCCHN = squamous cell carcinoma of the head and neck; SD = standard deviation; TNM = tumour, node and metastasis; VAS = visual analogue scale; WHO = World Health Organization; wt/vol = weight/volume.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Anné 2002

Not an RCT ‐ compares a subcutaneous amifostine group with the intravenous amifostine and no‐treatment arms from another study (Brizel 2000)

Bagga 2007

Abstract; insufficient information

Bakowski 1978

CCT

Belcaro 2008

Not head and neck cancer patients

Bohuslavizki 1998

Radioactive iodine not radiotherapy

Borg 2007

Abstract; insufficient information

Bourhis 2000

Salivary gland dysfunction was not a reported outcome

Braaksma 2002

Data unavailable for the outcomes of interest to this review

Braaksma 2005

Salivary gland dysfunction was not a reported outcome

Chambers 2005

Abstract; insufficient information

Demiroz 2012

Not an RCT

Fallahi 2013

Radioactive iodine not radiotherapy

Fan 2011

Unclear whether this is an RCT or not. Study authors contacted February 2016 but no reply received

Franzén 1995

Salivary gland dysfunction was not a reported outcome

Fuertes 2004

Quasi‐randomised trial (case history number)

Goyal 2007

Abstract; insufficient information

Gu 2014

Abstract; insufficient information

Johnson 2002

Not an RCT

Karacetin 2004

Quasi‐randomised trial

Koukourakis 2000

No formal assessment of salivary gland dysfunction; data not presented for head and neck cancer patients

Kumarchandra 2010

Abstract; insufficient information

Manoor 2014

Abstract; insufficient information

Mateos 2001

Quasi‐randomised trial (alternate assignment)

Mitine 2000

Abstract; insufficient information

Mix 2013

Abstract; insufficient information

Nicolatou‐Galitis 2003

Not an RCT

Norberg‐Spaak 1996

Abstract; insufficient information

Norberg‐Spaak 1997

Abstract; insufficient information

Nyárády 2006

Quasi‐randomised trial (alternate assignment)

Park 2012

Abstract; insufficient information

Park 2012a

Abstract; insufficient information

Peters 1999

Quasi‐randomised trial (day of birth)

Qian 2003

Dissertation; unable to obtain a copy

Resubal 2011

Abstract; insufficient information

Rieger 2012

Control group not relevant for this review

Rischin 2010

Tirapazamine is cancer treatment drug not a radiation protector

Rudat 2005

Abstract; insufficient information

Schönekäs 1999

Not an RCT

Sharma 2012

Intervention not defined as a pharmacological agent

Strnad 1997

Abstract; insufficient information

Su 2006

Salivary gland dysfunction measured as an adverse event following administration of G‐CSF

Takahashi 1986

Not an RCT

Thorstad 2003

Not an RCT

Uchiyama 2005

Unclear whether this is an RCT or not

Zale 1993

Abstract; insufficient information

Zimmerman 1997

Not an RCT

CCT = controlled clinical trial; G‐CSF = granulocyte‐colony stimulating factor; RCT = randomised controlled trial.

Characteristics of studies awaiting assessment [ordered by study ID]

Yu 2009

Methods

RCT

Participants

Nasopharynx cancer patients with poorly differentiated squamous carcinoma, at clinical phase III to VI, aged 25 to 69 years, first session of treatment, radiation field included > 75% of the area of salivary glands

Interventions

Amifostine versus no intervention

Outcomes

Xerostomia; salivary flow rates; adverse effects

Notes

Method/scale used to assess xerostomia unclear and further information required from study authors; the results do not have the potential to change any conclusions of the review

RCT = randomised controlled trial.

Characteristics of ongoing studies [ordered by study ID]

NCT02430298

Trial name or title

Topical and oral melatonin for preventing concurrent radiochemotherapy induced oral mucositis and xerostomia in head and neck cancer patients

Methods

Parallel, double‐blind RCT

Participants

Head and neck cancer adult patients

Interventions

Topical and oral melatonin versus placebo

Outcomes

Xerostomia; oral mucositis; quality of life

Starting date

July 2013

Contact information

Nutjaree Pratheepawanit Johns, Khon Kaen University

Notes

clinicaltrials.gov/show/NCT02430298

RCT = randomised controlled trial.

Data and analyses

Open in table viewer
Comparison 1. Pilocarpine versus no treatment/placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia Show forest plot

6

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 1 Xerostomia.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 1 Xerostomia.

1.1 End of radiotherapy

4

122

Std. Mean Difference (IV, Random, 95% CI)

0.20 [‐0.16, 0.56]

1.2 Up to and including 3 months postradiotherapy

3

125

Std. Mean Difference (IV, Random, 95% CI)

0.02 [‐0.33, 0.37]

1.3 Up to and including 6 months postradiotherapy

2

126

Std. Mean Difference (IV, Random, 95% CI)

‐0.35 [‐1.04, 0.33]

2 Xerostomia (LENT‐SOMA scale) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 1.2

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 2 Xerostomia (LENT‐SOMA scale).

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 2 Xerostomia (LENT‐SOMA scale).

2.1 Up to and including 6 months postradiotherapy

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

3 Xerostomia Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 1.3

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 3 Xerostomia.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 3 Xerostomia.

3.1 End of radiotherapy

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.2 Up to and including 3 months postradiotherapy

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Salivary flow rate (unstimulated) Show forest plot

3

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 4 Salivary flow rate (unstimulated).

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 4 Salivary flow rate (unstimulated).

4.1 End of radiotherapy

3

76

Std. Mean Difference (IV, Random, 95% CI)

0.24 [‐0.24, 0.72]

4.2 Up to and including 3 months postradiotherapy

1

24

Std. Mean Difference (IV, Random, 95% CI)

‐0.27 [‐1.07, 0.54]

5 Salivary flow rate (stimulated) Show forest plot

3

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 5 Salivary flow rate (stimulated).

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 5 Salivary flow rate (stimulated).

5.1 End of radiotherapy

2

58

Std. Mean Difference (IV, Random, 95% CI)

0.08 [‐0.44, 0.59]

5.2 Up to and including 3 months postradiotherapy

1

24

Std. Mean Difference (IV, Random, 95% CI)

0.40 [‐0.41, 1.21]

5.3 Up to and including 6 months postradiotherapy

1

9

Std. Mean Difference (IV, Random, 95% CI)

0.52 [‐0.84, 1.87]

5.4 Up to and including 12 months postradiotherapy

1

9

Std. Mean Difference (IV, Random, 95% CI)

0.53 [‐0.83, 1.88]

6 Salivary flow rate (> 0 g) unstimulated Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.6

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 6 Salivary flow rate (> 0 g) unstimulated.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 6 Salivary flow rate (> 0 g) unstimulated.

6.1 End of radiotherapy

1

154

Risk Ratio (M‐H, Random, 95% CI)

1.90 [0.98, 3.69]

6.2 Up to and including 3 months postradiotherapy

1

152

Risk Ratio (M‐H, Random, 95% CI)

2.0 [0.86, 4.68]

7 Salivary flow rate (> 0 g) stimulated Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.7

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 7 Salivary flow rate (> 0 g) stimulated.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 7 Salivary flow rate (> 0 g) stimulated.

7.1 End of radiotherapy

1

138

Risk Ratio (M‐H, Random, 95% CI)

1.87 [0.77, 4.52]

7.2 Up to and including 3 months postradiotherapy

1

139

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.23, 2.11]

8 Overall survival Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 1.8

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 8 Overall survival.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 8 Overall survival.

9 Quality of life Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 1.9

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 9 Quality of life.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 9 Quality of life.

9.1 End of radiotherapy

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

9.2 Up to and including 3 months postradiotherapy

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

9.3 Up to and including 6 months postradiotherapy

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 2. Biperiden plus pilocarpine versus no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Salivary flow rate (unstimulated) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 2.1

Comparison 2 Biperiden plus pilocarpine versus no treatment, Outcome 1 Salivary flow rate (unstimulated).

Comparison 2 Biperiden plus pilocarpine versus no treatment, Outcome 1 Salivary flow rate (unstimulated).

1.1 End of radiotherapy

1

40

Mean Difference (IV, Random, 95% CI)

0.02 [‐0.08, 0.12]

1.2 Up to and including 3 months postradiotherapy

1

17

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

2 Salivary flow rate (> 0 g) unstimulated Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.2

Comparison 2 Biperiden plus pilocarpine versus no treatment, Outcome 2 Salivary flow rate (> 0 g) unstimulated.

Comparison 2 Biperiden plus pilocarpine versus no treatment, Outcome 2 Salivary flow rate (> 0 g) unstimulated.

2.1 End of radiotherapy

1

60

Risk Ratio (M‐H, Random, 95% CI)

1.11 [0.77, 1.58]

2.2 Up to and including 3 months postradiotherapy

1

60

Risk Ratio (M‐H, Random, 95% CI)

7.50 [1.88, 29.99]

2.3 Up to and including 6 months postradiotherapy

1

60

Risk Ratio (M‐H, Random, 95% CI)

29.00 [1.81, 465.07]

2.4 Up to and including 12 months postradiotherapy

1

60

Risk Ratio (M‐H, Random, 95% CI)

35.00 [2.20, 556.71]

Open in table viewer
Comparison 3. Amifostine versus no treatment/placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (0 to 4 scale ‐ grade 2 or above) Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.1

Comparison 3 Amifostine versus no treatment/placebo, Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

Comparison 3 Amifostine versus no treatment/placebo, Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

1.1 End of radiotherapy

3

119

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.19, 0.67]

1.2 Up to and including 3 months postradiotherapy

5

687

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.48, 0.92]

1.3 12 months postradiotherapy

7

682

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.40, 1.23]

2 Salivary flow rate (unstimulated) Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 3.2

Comparison 3 Amifostine versus no treatment/placebo, Outcome 2 Salivary flow rate (unstimulated).

Comparison 3 Amifostine versus no treatment/placebo, Outcome 2 Salivary flow rate (unstimulated).

2.1 End of radiotherapy

2

83

Mean Difference (IV, Random, 95% CI)

0.34 [0.07, 0.61]

2.2 Up to and including 3 months postradiotherapy

1

41

Mean Difference (IV, Random, 95% CI)

0.13 [‐0.90, 1.16]

2.3 12 months postradiotherapy

1

27

Mean Difference (IV, Random, 95% CI)

0.32 [0.09, 0.55]

3 Salivary flow rate (unstimulated) ‐ incidence of > 0.1 g in 5 min Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.3

Comparison 3 Amifostine versus no treatment/placebo, Outcome 3 Salivary flow rate (unstimulated) ‐ incidence of > 0.1 g in 5 min.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 3 Salivary flow rate (unstimulated) ‐ incidence of > 0.1 g in 5 min.

3.1 12 months postradiotherapy

1

175

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.13, 1.86]

4 Salivary flow rate (stimulated) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 3.4

Comparison 3 Amifostine versus no treatment/placebo, Outcome 4 Salivary flow rate (stimulated).

Comparison 3 Amifostine versus no treatment/placebo, Outcome 4 Salivary flow rate (stimulated).

4.1 End of radiotherapy

1

47

Mean Difference (IV, Random, 95% CI)

‐0.09 [‐1.48, 1.30]

4.2 Up to and including 3 months postradiotherapy

1

41

Mean Difference (IV, Random, 95% CI)

0.38 [‐1.43, 2.19]

4.3 12 months postradiotherapy

1

27

Mean Difference (IV, Random, 95% CI)

0.82 [‐0.47, 2.11]

5 Salivary flow rate (stimulated) ‐ incidence of > 0.1 g in 5 min Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.5

Comparison 3 Amifostine versus no treatment/placebo, Outcome 5 Salivary flow rate (stimulated) ‐ incidence of > 0.1 g in 5 min.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 5 Salivary flow rate (stimulated) ‐ incidence of > 0.1 g in 5 min.

5.1 12 months postradiotherapy

1

173

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.89, 1.41]

6 Overall survival at 12 to 24 months postradiotherapy Show forest plot

2

271

Hazard ratio (Random, 95% CI)

1.18 [0.85, 1.66]

Analysis 3.6

Comparison 3 Amifostine versus no treatment/placebo, Outcome 6 Overall survival at 12 to 24 months postradiotherapy.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 6 Overall survival at 12 to 24 months postradiotherapy.

7 Overall survival ‐ narrative data Show forest plot

Other data

No numeric data

Analysis 3.7

Study

Time point

Amifostine

Control

Comments

Haddad 2009

Median follow‐up 34 months after radiotherapy, minimum 26 months

"No differences noted"

Jellema 2006

24 months

3 times weekly = 84%

5 times weekly = 58%

70%

Reported narratively rather than as a risk ratio due to differing results in the amifostine arms



Comparison 3 Amifostine versus no treatment/placebo, Outcome 7 Overall survival ‐ narrative data.

8 Progression‐free survival at 12 to 24 months postradiotherapy Show forest plot

2

247

Hazard ratio (Random, 95% CI)

0.94 [0.70, 1.27]

Analysis 3.8

Comparison 3 Amifostine versus no treatment/placebo, Outcome 8 Progression‐free survival at 12 to 24 months postradiotherapy.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 8 Progression‐free survival at 12 to 24 months postradiotherapy.

9 Progression‐free survival Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.9

Comparison 3 Amifostine versus no treatment/placebo, Outcome 9 Progression‐free survival.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 9 Progression‐free survival.

9.1 18 months postradiotherapy

1

45

Risk Ratio (M‐H, Random, 95% CI)

1.11 [0.81, 1.51]

10 Progression‐free survival ‐ narrative data Show forest plot

Other data

No numeric data

Analysis 3.10

Study

Time point

Amifostine

Control

Comments

Haddad 2009

Median follow‐up 34 months after radiotherapy, minimum 26 months

"No differences noted"



Comparison 3 Amifostine versus no treatment/placebo, Outcome 10 Progression‐free survival ‐ narrative data.

11 Locoregional tumour control at 12 to 24 months postradiotherapy Show forest plot

2

279

Hazard ratio (Random, 95% CI)

0.90 [0.74, 1.11]

Analysis 3.11

Comparison 3 Amifostine versus no treatment/placebo, Outcome 11 Locoregional tumour control at 12 to 24 months postradiotherapy.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 11 Locoregional tumour control at 12 to 24 months postradiotherapy.

12 Locoregional tumour control ‐ narrative data Show forest plot

Other data

No numeric data

Analysis 3.12

Study

Time point

Amifostine

Control

Comments

Haddad 2009

Median follow‐up 34 months after radiotherapy, minimum 26 months

"No differences noted"

Jellema 2006

24 months

3 times weekly = 67%

5 times weekly = 83%

79%

Reported narratively rather than as a risk ratio due to differing results in the amifostine arms

Patni 2004

24 month

No data

No data

"Amifostine does not alter the response or the survival"



Comparison 3 Amifostine versus no treatment/placebo, Outcome 12 Locoregional tumour control ‐ narrative data.

13 Disease‐free survival Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.13

Comparison 3 Amifostine versus no treatment/placebo, Outcome 13 Disease‐free survival.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 13 Disease‐free survival.

13.1 24 months postradiotherapy

1

170

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.73, 1.21]

14 Disease‐free survival Show forest plot

Other data

No numeric data

Analysis 3.14

Study

Time point

Amifostine

Control

Comments

Patni 2004

24 months

No data

No data

"Amifostine does not alter the response or the survival"

Veerasarn 2006

24 months

No data

No data

"There was no statistical difference in 2‐year disease‐free survival"



Comparison 3 Amifostine versus no treatment/placebo, Outcome 14 Disease‐free survival.

15 Quality of life (Patient Benefit Questionnaire) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 3.15

Comparison 3 Amifostine versus no treatment/placebo, Outcome 15 Quality of life (Patient Benefit Questionnaire).

Comparison 3 Amifostine versus no treatment/placebo, Outcome 15 Quality of life (Patient Benefit Questionnaire).

15.1 End of radiotherapy

1

298

Mean Difference (IV, Random, 95% CI)

0.38 [‐0.07, 0.83]

15.2 Up to and including 3 months postradiotherapy

1

233

Mean Difference (IV, Random, 95% CI)

0.52 [‐0.02, 1.06]

15.3 12 months postradiotherapy

1

180

Mean Difference (IV, Random, 95% CI)

0.70 [0.20, 1.20]

Open in table viewer
Comparison 4. Amifostine (comparison of dosages)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (0 to 4 scale ‐ grade 2 or above) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.1

Comparison 4 Amifostine (comparison of dosages), Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

Comparison 4 Amifostine (comparison of dosages), Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

1.1 12 months postradiotherapy

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.58, 1.53]

2 Overall survival ‐ narrative data Show forest plot

Other data

No numeric data

Analysis 4.2

Study

Time point

Amifostine 3 times weekly

Amifostine 5 times weekly

Comments

Jellema 2006

24 months

84%

58%



Comparison 4 Amifostine (comparison of dosages), Outcome 2 Overall survival ‐ narrative data.

3 Locoregional tumour control ‐ narrative data Show forest plot

Other data

No numeric data

Analysis 4.3

Study

Time point

Amifostine 3 times weekly

Amifostine 5 times weekly

Comments

Jellema 2006

24 months

67%

83%



Comparison 4 Amifostine (comparison of dosages), Outcome 3 Locoregional tumour control ‐ narrative data.

Open in table viewer
Comparison 5. Amifostine (intravenous versus subcutaneous)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (0 to 4 scale ‐ grade 2 or above) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.1

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

1.1 Up to and including 3 months postradiotherapy

1

263

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.76, 1.40]

1.2 12 months postradiotherapy

1

127

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.42, 0.88]

2 Overall survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

Analysis 5.2

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 2 Overall survival.

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 2 Overall survival.

2.1 48 months after radiotherapy

1

Hazard Ratio (Random, 95% CI)

1.36 [0.89, 2.10]

3 Locoregional tumour control Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

Analysis 5.3

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 3 Locoregional tumour control.

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 3 Locoregional tumour control.

3.1 48 months after radiotherapy

1

Hazard Ratio (Random, 95% CI)

1.34 [0.76, 2.36]

Open in table viewer
Comparison 6. Chinese medicine versus no treatment/placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 6.1

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 1 Xerostomia.

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 1 Xerostomia.

1.1 End of radiotherapy: Shenqi Fanghou recipe versus no intervention

1

140

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.28, 0.55]

1.2 End of radiotherapy: TWBXM versus placebo

1

71

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.78, 1.03]

1.3 Up to and including 3 months postradiotherapy: Jinlong capsules versus no intervention

1

95

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.59, 1.36]

2 Xerostomia Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.2

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 2 Xerostomia.

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 2 Xerostomia.

2.1 End of radiotherapy: TWBXM versus placebo

1

68

Mean Difference (IV, Random, 95% CI)

‐2.41 [‐16.19, 11.37]

2.2 Up to and including 3 months postradiotherapy: TWBXM versus placebo

1

44

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐17.21, 17.01]

3 Salivary flow rate (stimulated) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.3

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 3 Salivary flow rate (stimulated).

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 3 Salivary flow rate (stimulated).

3.1 End of radiotherapy: Chinese medicine versus no intervention

1

50

Mean Difference (IV, Random, 95% CI)

0.09 [0.03, 0.15]

4 Overall survival (12 months postRT) Show forest plot

1

78

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.84, 1.30]

Analysis 6.4

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 4 Overall survival (12 months postRT).

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 4 Overall survival (12 months postRT).

5 Quality of life (EORTC‐C30) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.5

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 5 Quality of life (EORTC‐C30).

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 5 Quality of life (EORTC‐C30).

5.1 End of radiotherapy: TWBXM versus placebo

1

68

Mean Difference (IV, Random, 95% CI)

2.39 [‐8.74, 13.52]

5.2 Up to and including 3 months postradiotherapy: TWBXM versus placebo

1

44

Mean Difference (IV, Random, 95% CI)

1.93 [‐13.04, 16.90]

Open in table viewer
Comparison 7. Palifermin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (0 to 4 scale ‐ grade 2 or above) Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 7.1

Comparison 7 Palifermin versus placebo, Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

Comparison 7 Palifermin versus placebo, Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

1.1 Up to and including 3 months postRT

3

471

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.77, 1.22]

2 Overall survival at 42 to 72 months from baseline Show forest plot

3

Hazard Ratio (Random, 95% CI)

1.00 [0.72, 1.39]

Analysis 7.2

Comparison 7 Palifermin versus placebo, Outcome 2 Overall survival at 42 to 72 months from baseline.

Comparison 7 Palifermin versus placebo, Outcome 2 Overall survival at 42 to 72 months from baseline.

3 Progression‐free survival at 42 to 72 months from baseline Show forest plot

3

Hazard Ratio (Random, 95% CI)

1.06 [0.80, 1.42]

Analysis 7.3

Comparison 7 Palifermin versus placebo, Outcome 3 Progression‐free survival at 42 to 72 months from baseline.

Comparison 7 Palifermin versus placebo, Outcome 3 Progression‐free survival at 42 to 72 months from baseline.

Open in table viewer
Comparison 8. Bethanechol versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (0 to 3 scale ‐ grade 2 or above) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 8.1

Comparison 8 Bethanechol versus placebo, Outcome 1 Xerostomia (0 to 3 scale ‐ grade 2 or above).

Comparison 8 Bethanechol versus placebo, Outcome 1 Xerostomia (0 to 3 scale ‐ grade 2 or above).

1.1 End of radiotherapy

1

84

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.28, 0.66]

1.2 Up to and including 3 months postradiotherapy

1

84

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.65, 1.01]

2 Salivary flow rate (unstimulated) ‐ ml/min Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 8.2

Comparison 8 Bethanechol versus placebo, Outcome 2 Salivary flow rate (unstimulated) ‐ ml/min.

Comparison 8 Bethanechol versus placebo, Outcome 2 Salivary flow rate (unstimulated) ‐ ml/min.

2.1 2 months postradiotherapy

1

97

Mean Difference (IV, Random, 95% CI)

0.19 [0.06, 0.32]

3 Salivary flow rate (stimulated) ‐ ml/min Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 8.3

Comparison 8 Bethanechol versus placebo, Outcome 3 Salivary flow rate (stimulated) ‐ ml/min.

Comparison 8 Bethanechol versus placebo, Outcome 3 Salivary flow rate (stimulated) ‐ ml/min.

3.1 2 months postradiotherapy

1

97

Mean Difference (IV, Random, 95% CI)

0.15 [‐0.03, 0.33]

Open in table viewer
Comparison 9. Bethanechol versus artificial saliva

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (dry mouth? yes/no) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 9.1

Comparison 9 Bethanechol versus artificial saliva, Outcome 1 Xerostomia (dry mouth? yes/no).

Comparison 9 Bethanechol versus artificial saliva, Outcome 1 Xerostomia (dry mouth? yes/no).

1.1 End of radiotherapy

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.30, 1.29]

1.2 8 to 40 weeks postradiotherapy

1

30

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.30, 1.05]

2 Salivary flow rate (unstimulated) ‐ ml/min Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 9.2

Comparison 9 Bethanechol versus artificial saliva, Outcome 2 Salivary flow rate (unstimulated) ‐ ml/min.

Comparison 9 Bethanechol versus artificial saliva, Outcome 2 Salivary flow rate (unstimulated) ‐ ml/min.

2.1 End of radiotherapy

1

36

Mean Difference (IV, Random, 95% CI)

0.12 [0.01, 0.23]

2.2 8 to 40 weeks postradiotherapy

1

33

Mean Difference (IV, Random, 95% CI)

0.07 [‐0.02, 0.16]

3 Salivary flow rate (stimulated) ‐ ml/min Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 9.3

Comparison 9 Bethanechol versus artificial saliva, Outcome 3 Salivary flow rate (stimulated) ‐ ml/min.

Comparison 9 Bethanechol versus artificial saliva, Outcome 3 Salivary flow rate (stimulated) ‐ ml/min.

3.1 End of radiotherapy

1

32

Mean Difference (IV, Random, 95% CI)

0.13 [‐0.03, 0.29]

3.2 8 to 40 weeks postradiotherapy

1

29

Mean Difference (IV, Random, 95% CI)

0.21 [0.01, 0.41]

4 Overall survival Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 9.4

Comparison 9 Bethanechol versus artificial saliva, Outcome 4 Overall survival.

Comparison 9 Bethanechol versus artificial saliva, Outcome 4 Overall survival.

4.1 40 weeks postradiotherapy

1

43

Risk Ratio (M‐H, Random, 95% CI)

1.59 [0.43, 5.84]

Open in table viewer
Comparison 10. Selenium versus no selenium

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia Show forest plot

Other data

No numeric data

Analysis 10.1

Study

Büntzel 2010

"comparing the mean value of xerostomia, no statistically significant difference can be seen between the groups"



Comparison 10 Selenium versus no selenium, Outcome 1 Xerostomia.

Open in table viewer
Comparison 11. Antimicrobial lozenge versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (QoL response for dryness) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 11.1

Comparison 11 Antimicrobial lozenge versus placebo, Outcome 1 Xerostomia (QoL response for dryness).

Comparison 11 Antimicrobial lozenge versus placebo, Outcome 1 Xerostomia (QoL response for dryness).

1.1 Up to and including 3 months postradiotherapy

1

133

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.97, 1.40]

2 Quality of life Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 11.2

Comparison 11 Antimicrobial lozenge versus placebo, Outcome 2 Quality of life.

Comparison 11 Antimicrobial lozenge versus placebo, Outcome 2 Quality of life.

2.1 Up to and including 3 months postradiotherapy (change score over 6 months)

1

131

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.65, 1.50]

Open in table viewer
Comparison 12. Polaprezinc versus azulene oral rinse

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (grade 2 or above) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 12.1

Comparison 12 Polaprezinc versus azulene oral rinse, Outcome 1 Xerostomia (grade 2 or above).

Comparison 12 Polaprezinc versus azulene oral rinse, Outcome 1 Xerostomia (grade 2 or above).

1.1 End of radiotherapy

1

31

Risk Ratio (M‐H, Random, 95% CI)

0.17 [0.04, 0.65]

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

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

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 3

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

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 1 Xerostomia.
Figuras y tablas -
Analysis 1.1

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 1 Xerostomia.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 2 Xerostomia (LENT‐SOMA scale).
Figuras y tablas -
Analysis 1.2

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 2 Xerostomia (LENT‐SOMA scale).

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 3 Xerostomia.
Figuras y tablas -
Analysis 1.3

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 3 Xerostomia.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 4 Salivary flow rate (unstimulated).
Figuras y tablas -
Analysis 1.4

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 4 Salivary flow rate (unstimulated).

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 5 Salivary flow rate (stimulated).
Figuras y tablas -
Analysis 1.5

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 5 Salivary flow rate (stimulated).

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 6 Salivary flow rate (> 0 g) unstimulated.
Figuras y tablas -
Analysis 1.6

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 6 Salivary flow rate (> 0 g) unstimulated.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 7 Salivary flow rate (> 0 g) stimulated.
Figuras y tablas -
Analysis 1.7

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 7 Salivary flow rate (> 0 g) stimulated.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 8 Overall survival.
Figuras y tablas -
Analysis 1.8

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 8 Overall survival.

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 9 Quality of life.
Figuras y tablas -
Analysis 1.9

Comparison 1 Pilocarpine versus no treatment/placebo, Outcome 9 Quality of life.

Comparison 2 Biperiden plus pilocarpine versus no treatment, Outcome 1 Salivary flow rate (unstimulated).
Figuras y tablas -
Analysis 2.1

Comparison 2 Biperiden plus pilocarpine versus no treatment, Outcome 1 Salivary flow rate (unstimulated).

Comparison 2 Biperiden plus pilocarpine versus no treatment, Outcome 2 Salivary flow rate (> 0 g) unstimulated.
Figuras y tablas -
Analysis 2.2

Comparison 2 Biperiden plus pilocarpine versus no treatment, Outcome 2 Salivary flow rate (> 0 g) unstimulated.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).
Figuras y tablas -
Analysis 3.1

Comparison 3 Amifostine versus no treatment/placebo, Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

Comparison 3 Amifostine versus no treatment/placebo, Outcome 2 Salivary flow rate (unstimulated).
Figuras y tablas -
Analysis 3.2

Comparison 3 Amifostine versus no treatment/placebo, Outcome 2 Salivary flow rate (unstimulated).

Comparison 3 Amifostine versus no treatment/placebo, Outcome 3 Salivary flow rate (unstimulated) ‐ incidence of > 0.1 g in 5 min.
Figuras y tablas -
Analysis 3.3

Comparison 3 Amifostine versus no treatment/placebo, Outcome 3 Salivary flow rate (unstimulated) ‐ incidence of > 0.1 g in 5 min.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 4 Salivary flow rate (stimulated).
Figuras y tablas -
Analysis 3.4

Comparison 3 Amifostine versus no treatment/placebo, Outcome 4 Salivary flow rate (stimulated).

Comparison 3 Amifostine versus no treatment/placebo, Outcome 5 Salivary flow rate (stimulated) ‐ incidence of > 0.1 g in 5 min.
Figuras y tablas -
Analysis 3.5

Comparison 3 Amifostine versus no treatment/placebo, Outcome 5 Salivary flow rate (stimulated) ‐ incidence of > 0.1 g in 5 min.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 6 Overall survival at 12 to 24 months postradiotherapy.
Figuras y tablas -
Analysis 3.6

Comparison 3 Amifostine versus no treatment/placebo, Outcome 6 Overall survival at 12 to 24 months postradiotherapy.

Study

Time point

Amifostine

Control

Comments

Haddad 2009

Median follow‐up 34 months after radiotherapy, minimum 26 months

"No differences noted"

Jellema 2006

24 months

3 times weekly = 84%

5 times weekly = 58%

70%

Reported narratively rather than as a risk ratio due to differing results in the amifostine arms

Figuras y tablas -
Analysis 3.7

Comparison 3 Amifostine versus no treatment/placebo, Outcome 7 Overall survival ‐ narrative data.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 8 Progression‐free survival at 12 to 24 months postradiotherapy.
Figuras y tablas -
Analysis 3.8

Comparison 3 Amifostine versus no treatment/placebo, Outcome 8 Progression‐free survival at 12 to 24 months postradiotherapy.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 9 Progression‐free survival.
Figuras y tablas -
Analysis 3.9

Comparison 3 Amifostine versus no treatment/placebo, Outcome 9 Progression‐free survival.

Study

Time point

Amifostine

Control

Comments

Haddad 2009

Median follow‐up 34 months after radiotherapy, minimum 26 months

"No differences noted"

Figuras y tablas -
Analysis 3.10

Comparison 3 Amifostine versus no treatment/placebo, Outcome 10 Progression‐free survival ‐ narrative data.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 11 Locoregional tumour control at 12 to 24 months postradiotherapy.
Figuras y tablas -
Analysis 3.11

Comparison 3 Amifostine versus no treatment/placebo, Outcome 11 Locoregional tumour control at 12 to 24 months postradiotherapy.

Study

Time point

Amifostine

Control

Comments

Haddad 2009

Median follow‐up 34 months after radiotherapy, minimum 26 months

"No differences noted"

Jellema 2006

24 months

3 times weekly = 67%

5 times weekly = 83%

79%

Reported narratively rather than as a risk ratio due to differing results in the amifostine arms

Patni 2004

24 month

No data

No data

"Amifostine does not alter the response or the survival"

Figuras y tablas -
Analysis 3.12

Comparison 3 Amifostine versus no treatment/placebo, Outcome 12 Locoregional tumour control ‐ narrative data.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 13 Disease‐free survival.
Figuras y tablas -
Analysis 3.13

Comparison 3 Amifostine versus no treatment/placebo, Outcome 13 Disease‐free survival.

Study

Time point

Amifostine

Control

Comments

Patni 2004

24 months

No data

No data

"Amifostine does not alter the response or the survival"

Veerasarn 2006

24 months

No data

No data

"There was no statistical difference in 2‐year disease‐free survival"

Figuras y tablas -
Analysis 3.14

Comparison 3 Amifostine versus no treatment/placebo, Outcome 14 Disease‐free survival.

Comparison 3 Amifostine versus no treatment/placebo, Outcome 15 Quality of life (Patient Benefit Questionnaire).
Figuras y tablas -
Analysis 3.15

Comparison 3 Amifostine versus no treatment/placebo, Outcome 15 Quality of life (Patient Benefit Questionnaire).

Comparison 4 Amifostine (comparison of dosages), Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).
Figuras y tablas -
Analysis 4.1

Comparison 4 Amifostine (comparison of dosages), Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

Study

Time point

Amifostine 3 times weekly

Amifostine 5 times weekly

Comments

Jellema 2006

24 months

84%

58%

Figuras y tablas -
Analysis 4.2

Comparison 4 Amifostine (comparison of dosages), Outcome 2 Overall survival ‐ narrative data.

Study

Time point

Amifostine 3 times weekly

Amifostine 5 times weekly

Comments

Jellema 2006

24 months

67%

83%

Figuras y tablas -
Analysis 4.3

Comparison 4 Amifostine (comparison of dosages), Outcome 3 Locoregional tumour control ‐ narrative data.

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).
Figuras y tablas -
Analysis 5.1

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 2 Overall survival.
Figuras y tablas -
Analysis 5.2

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 2 Overall survival.

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 3 Locoregional tumour control.
Figuras y tablas -
Analysis 5.3

Comparison 5 Amifostine (intravenous versus subcutaneous), Outcome 3 Locoregional tumour control.

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 1 Xerostomia.
Figuras y tablas -
Analysis 6.1

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 1 Xerostomia.

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 2 Xerostomia.
Figuras y tablas -
Analysis 6.2

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 2 Xerostomia.

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 3 Salivary flow rate (stimulated).
Figuras y tablas -
Analysis 6.3

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 3 Salivary flow rate (stimulated).

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 4 Overall survival (12 months postRT).
Figuras y tablas -
Analysis 6.4

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 4 Overall survival (12 months postRT).

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 5 Quality of life (EORTC‐C30).
Figuras y tablas -
Analysis 6.5

Comparison 6 Chinese medicine versus no treatment/placebo, Outcome 5 Quality of life (EORTC‐C30).

Comparison 7 Palifermin versus placebo, Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).
Figuras y tablas -
Analysis 7.1

Comparison 7 Palifermin versus placebo, Outcome 1 Xerostomia (0 to 4 scale ‐ grade 2 or above).

Comparison 7 Palifermin versus placebo, Outcome 2 Overall survival at 42 to 72 months from baseline.
Figuras y tablas -
Analysis 7.2

Comparison 7 Palifermin versus placebo, Outcome 2 Overall survival at 42 to 72 months from baseline.

Comparison 7 Palifermin versus placebo, Outcome 3 Progression‐free survival at 42 to 72 months from baseline.
Figuras y tablas -
Analysis 7.3

Comparison 7 Palifermin versus placebo, Outcome 3 Progression‐free survival at 42 to 72 months from baseline.

Comparison 8 Bethanechol versus placebo, Outcome 1 Xerostomia (0 to 3 scale ‐ grade 2 or above).
Figuras y tablas -
Analysis 8.1

Comparison 8 Bethanechol versus placebo, Outcome 1 Xerostomia (0 to 3 scale ‐ grade 2 or above).

Comparison 8 Bethanechol versus placebo, Outcome 2 Salivary flow rate (unstimulated) ‐ ml/min.
Figuras y tablas -
Analysis 8.2

Comparison 8 Bethanechol versus placebo, Outcome 2 Salivary flow rate (unstimulated) ‐ ml/min.

Comparison 8 Bethanechol versus placebo, Outcome 3 Salivary flow rate (stimulated) ‐ ml/min.
Figuras y tablas -
Analysis 8.3

Comparison 8 Bethanechol versus placebo, Outcome 3 Salivary flow rate (stimulated) ‐ ml/min.

Comparison 9 Bethanechol versus artificial saliva, Outcome 1 Xerostomia (dry mouth? yes/no).
Figuras y tablas -
Analysis 9.1

Comparison 9 Bethanechol versus artificial saliva, Outcome 1 Xerostomia (dry mouth? yes/no).

Comparison 9 Bethanechol versus artificial saliva, Outcome 2 Salivary flow rate (unstimulated) ‐ ml/min.
Figuras y tablas -
Analysis 9.2

Comparison 9 Bethanechol versus artificial saliva, Outcome 2 Salivary flow rate (unstimulated) ‐ ml/min.

Comparison 9 Bethanechol versus artificial saliva, Outcome 3 Salivary flow rate (stimulated) ‐ ml/min.
Figuras y tablas -
Analysis 9.3

Comparison 9 Bethanechol versus artificial saliva, Outcome 3 Salivary flow rate (stimulated) ‐ ml/min.

Comparison 9 Bethanechol versus artificial saliva, Outcome 4 Overall survival.
Figuras y tablas -
Analysis 9.4

Comparison 9 Bethanechol versus artificial saliva, Outcome 4 Overall survival.

Study

Büntzel 2010

"comparing the mean value of xerostomia, no statistically significant difference can be seen between the groups"

Figuras y tablas -
Analysis 10.1

Comparison 10 Selenium versus no selenium, Outcome 1 Xerostomia.

Comparison 11 Antimicrobial lozenge versus placebo, Outcome 1 Xerostomia (QoL response for dryness).
Figuras y tablas -
Analysis 11.1

Comparison 11 Antimicrobial lozenge versus placebo, Outcome 1 Xerostomia (QoL response for dryness).

Comparison 11 Antimicrobial lozenge versus placebo, Outcome 2 Quality of life.
Figuras y tablas -
Analysis 11.2

Comparison 11 Antimicrobial lozenge versus placebo, Outcome 2 Quality of life.

Comparison 12 Polaprezinc versus azulene oral rinse, Outcome 1 Xerostomia (grade 2 or above).
Figuras y tablas -
Analysis 12.1

Comparison 12 Polaprezinc versus azulene oral rinse, Outcome 1 Xerostomia (grade 2 or above).

Summary of findings for the main comparison. Pilocarpine compared to no treatment/placebo for preventing salivary gland dysfunction following radiotherapy

Pilocarpine compared to no treatment/placebo for preventing salivary gland dysfunction following radiotherapy

Patient or population: patients receiving radiotherapy on its own or in addition to chemotherapy to the head and neck region
Intervention: pilocarpine
Comparison: no treatment/placebo

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Number of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with no treatment/placebo

Risk with pilocarpine

Xerostomia ‐ Up to and including 6 months postRT

Studies used different ways of measuring the outcome and therefore we combined the studies using SMD

SMD 0.35 lower
(1.04 lower to 0.33 higher)

126
(2 RCTs)

⊕⊝⊝⊝
VERY LOW1

Insufficient evidence of a difference at this time point and also at the end of RT and 3 months postRT

1 of the 2 studies in this assessment showed inconsistent results when using an alternative way of measuring this outcome at the 6‐month time point. 2 further studies showed insufficient evidence of a difference, 1 at the end of RT and the other at 3 months postRT

Salivary flow rate (unstimulated) ‐ Up to and including 3 months postRT

Studies used different ways of measuring the outcome and therefore we combined the studies using SMD

MD 0.06 lower (0.23 lower to 0.11 higher)

24
(1 RCT)

⊕⊝⊝⊝
VERY LOW2

Insufficient evidence of a difference at this time point and also at the end of RT

Same results for stimulated salivary flow rates at end of RT, and 3, 6 and 12 months postRT

Same results for a further study at the end of RT and 3 months postRT looking at whether or not stimulated and unstimulated salivary flow was > 0 g

Overall survival ‐ Up to and including 6 months postRT

724 per 1000

775 per 1000
(579 to 1000)

RR 1.07
(0.80 to 1.43)

60
(1 RCT)

⊕⊝⊝⊝
VERY LOW3

Insufficient evidence of a difference

Quality of life ‐ Up to and including 6 months postRT

McMaster University Head and Neck Questionnaire (HNRQ). Score 1‐7, lower score = poorer quality of life

Control group mean was 5.3

MD 0.20 higher
(0.19 lower to 0.59 higher)

90
(1 RCT)

⊕⊝⊝⊝
VERY LOW3

Insufficient evidence of a difference at this time point and also at the end of RT and 3 months postRT

Adverse effects

Insufficient evidence of a difference between groups for any reported adverse event, apart from for sweating where data from 5 studies showed an increased risk associated with pilocarpine (RR 2.98, 95% CI 1.43 to 6.22; P = 0.004; I2 = 0%; 389 participants; ⊕⊕⊝⊝ LOW4)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI)

CI: confidence interval; MD: mean difference; RCT: randomised controlled trial; RR: risk ratio; SMD: standardised mean difference; 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 1 level for risk of bias, 1 level for imprecision (small sample size and 95% CIs include both possibility of benefit and harm), and 1 level for inconsistency (I2 = 68%).
2Downgraded by 1 level for risk of bias, and 2 levels for imprecision (single study with 12 participants per group and 95% CIs include both possibility of benefit and harm).
3Downgraded by 1 level for risk of bias, and 2 levels for imprecision (single study and 95% CIs include both possibility of benefit and harm).
4Downgraded by 1 level for risk of bias, and 1 level for imprecision (very wide 95% CIs).

Figuras y tablas -
Summary of findings for the main comparison. Pilocarpine compared to no treatment/placebo for preventing salivary gland dysfunction following radiotherapy
Summary of findings 2. Amifostine compared to no treatment/placebo for preventing salivary gland dysfunction following radiotherapy

Amifostine compared to no treatment/placebo for preventing salivary gland dysfunction following radiotherapy

Patient or population: patients receiving radiotherapy on its own or in addition to chemotherapy to the head and neck region
Intervention: amifostine
Comparison: no treatment/placebo

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Number of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with no treatment/placebo

Risk with amifostine

Xerostomia (0‐4 scale ‐ grade 2 or above) ‐ 12 months postRT

418 per 1000

292 per 1000
(167 to 514)

RR 0.70
(0.40 to 1.23)

682
(7 studies)

⊕⊕⊝⊝
LOW1

Insufficient evidence of a difference at this time point. However, both at the end of RT (RR 0.35, 95% CI 0.19 to 0.67; 3 studies, 119 participants) and up to 3 months postRT (RR 0.66, 95% CI 0.48 to 0.92; 5 studies, 687 participants), amifostine reduced the risk of developing grade ≥ 2 xerostomia

Salivary flow rate (mg/5 min) (unstimulated) ‐ 12 months postRT

Control group mean was 0.16

MD 0.32 higher
(0.09 higher to 0.55 higher)

27
(1 study)

⊕⊝⊝⊝
VERY LOW2

Amifostine led to increased unstimulated saliva flow both at 12 months postRT and at the end of RT, but there was insufficient evidence of a difference at 3 months postRT. This evidence was supported by a further study showing a benefit for amifostine at 12 months postRT when looking at incidence of producing > 0.1 g of saliva over 5 minutes (RR 1.45, 95% CI 1.13 to 1.86; 175 participants). A further study narratively reported no difference

Insufficient evidence of a difference in stimulated saliva flow at any time point

Overall survival at 12 to 24 months postRT

450 per 1000**

531 per 1000
(383 to 747)

HR 1.18
(0.85 to 1.66)

271
(2 studies)

⊕⊝⊝⊝
VERY LOW3

Insufficient evidence to determine whether or not amifostine reduces overall survival, progression‐free survival, disease‐free survival or locoregional tumour control up to 24 months postRT

Quality of life (Patient Benefit Questionnaire) ‐ 12 months postRT

8 items each on a 10‐point scale where higher = better QoL

Control group mean was 6.66

MD 0.7 higher
(0.2 higher to 1.2 higher)

180
(1 study)

⊕⊝⊝⊝
VERY LOW2

Amifostine led to a small improvement in quality of life at 12 months postRT, but there was insufficient evidence of a difference at the end of RT and 3 months postRT

A further study narratively reported no difference at end of RT and 6, 12, 18, and 24 months postRT

Adverse effects

  • Data from 5 studies showed an increased risk of vomiting associated with amifostine (RR 4.90, 95% CI 2.87 to 8.38; 601 participants; ⊕⊕⊝⊝ LOW4)

  • Data from 3 studies showed an increased risk of hypotension associated with amifostine (RR 9.20, 95% CI 2.84 to 29.83; 376 participants; ⊕⊕⊝⊝ LOW4)

  • Data from 4 studies showed an increased risk of nausea associated with amifostine (RR 2.60, 95% CI 1.81 to 3.74; 556 participants; ⊕⊕⊝⊝ LOW4)

  • Data from 3 studies showed an increased risk of allergic response associated with amifostine (RR 7.51, 95% CI 1.40 to 40.39; 524 participants; ⊕⊕⊝⊝ LOW4)

There was insufficient evidence of a difference between groups for any other adverse events

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI)
**2014 5‐year overall survival rate of patients with head and neck squamous cell carcinoma (www.who.int/selection_medicines/committees/expert/20/applications/HeadNeck.pdf)

CI: confidence interval; HR: hazard ratio; MD: mean difference; QoL: quality of life; 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 1 level for risk of bias, and 1 level for inconsistency (I2 = 83%).
2Downgraded by 1 level for risk of bias, and 2 levels for imprecision (single study and small sample size).
3Downgraded by 1 level for risk of bias, and 2 levels for imprecision (small sample size and 95% CIs include both possibility of benefit and harm).
4Downgraded by 1 level for risk of bias, and 1 level for imprecision (very wide 95% CIs).

Figuras y tablas -
Summary of findings 2. Amifostine compared to no treatment/placebo for preventing salivary gland dysfunction following radiotherapy
Summary of findings 3. Palifermin compared to placebo for preventing salivary gland dysfunction following radiotherapy

Palifermin compared to placebo for preventing salivary gland dysfunction following radiotherapy

Patient or population: patients receiving radiotherapy on its own or in addition to chemotherapy to the head and neck region
Intervention: palifermin
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 palifermin

Xerostomia (0‐4 scale ‐ grade 2 or above) ‐ Up to and including 3 months postRT

727 per 1000

705 per 1000
(560 to 887)

RR 0.97
(0.77 to 1.22)

471
(3 studies)

⊕⊕⊝⊝
LOW1

Insufficient evidence of a difference at this time point

Overall survival at 42 to 72 months from baseline

450 per 1000**

450 per 1000
(324 to 626)

HR 1.00
(0.72 to 1.39)

(3 studies)

⊕⊕⊕⊝
MODERATE2

Insufficient evidence to determine whether or not amifostine reduces overall survival and progression‐free survival up to 72 months

Adverse effects

There was insufficient evidence of patients in either group experiencing more or less adverse events

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI)

**2014 5‐year overall survival rate of patients with head and neck squamous cell carcinoma (www.who.int/selection_medicines/committees/expert/20/applications/HeadNeck.pdf)

CI: confidence interval; HR: hazard ratio; 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 1 level for imprecision (95% CIs include both possibility of benefit and harm), and 1 level for inconsistency (I2 = 76%).
2Downgraded by 1 level for imprecision (95% CIs include both possibility of benefit and harm).

Figuras y tablas -
Summary of findings 3. Palifermin compared to placebo for preventing salivary gland dysfunction following radiotherapy
Table 1. Pilocarpine versus no treatment/placebo (other outcomes)

Outcome

Study ID

Time point

Pilocarpine

Control

Results

Comments

Oral related symptoms (other than salivary gland dysfunction/xerostomia)

Oral discomfort

Gornitsky 2004

End of radiotherapy

Mean 38.7 (SD 31.6)

n = 16

Mean 56.7 (SD 26.7)

n = 20

Mean difference ‐18.00 (95% CI ‐37.41 to 1.41), P = 0.07

Speech difficulties

Gornitsky 2004

Mean 57.5 (SD 26.8)

n = 16

Mean 37.3 (SD 27.5)

n = 18

Mean difference 20.20 (95% CI 1.93 to 38.47), P = 0.03

Eating difficulties

Gornitsky 2004

Mean 47.4 (SD 33.9)

n = 15

Mean 61.8 (SD 25.4)

n = 17

Mean difference ‐14.40 (95% CI ‐35.38 to 6.58), P = 0.18

Mucosal pain

Gornitsky 2004

Mean 38.8 (SD 33.9)

n = 17

Mean 53.6 (SD 34.2)

n = 19

Mean difference ‐14.80 (95% CI ‐37.07 to 7.47), P = 0.19

Oral complications

Pimentel 2014

1/5

4/6

RR 0.30 (95% CI 0.05 to 1.89), P = 0.20

Adverse events

Sweating

Abacioglu 1997

2/12

0/12

Random‐effects meta‐analysis of 5 studies: RR 2.98 (95% CI 1.43 to 6.22), P = 0.004

Heterogeneity: I2 = 0%, P = 0.52

Fisher 2003

18/118

5/114

Gornitsky 2004

3/28

1/28

Lozada‐Nur 1998

5/12

1/12

Sangthawan 2001

1/29

2/24

Chilling

Abacioglu 1997

1/12

0/12

RR 3.00 (95% CI 0.13 to 67.06), P = 0.49

Nausea

Gornitsky 2004

7/28

5/28

Random‐effects meta‐analysis of 3 studies: RR 1.39 (95% CI 0.63 to 3.05), P = 0.41

Heterogeneity: I2 = 0%, P = 0.93

Haddad 2002

3/18

3/21

Lozada‐Nur 1998

2/12

1/12

Vomiting

Fisher 2003

13/118

10/114

Random‐effects meta‐analysis of 3 studies: RR 1.28 (95% CI 0.70 to 2.35), P = 0.43

Heterogeneity: I2 = 0%, P = 0.92

Gornitsky 2004

6/28

5/28

Lozada‐Nur 1998

2/12

1/12

Headache

Gornitsky 2004

2/28

3/28

RR 0.67 (95% CI 0.12 to 3.69), P = 0.64

Excessive lacrimation (tears)

Fisher 2003

3/118

0/114

Random‐effects meta‐analysis of 3 studies: RR 2.54 (95% CI 0.70 to 9.17), P = 0.15

Heterogeneity: I2 = 0%, P = 0.71

Haddad 2002

1/18

0/21

Sangthawan 2001

4/25

2/22

Dysphasia

Lozada‐Nur 1998

3/12

2/12

RR 1.50 (95% CI 0.30 to 7.43), P = 0.62

Weakness

Fisher 2003

3/118

2/114

RR 1.45 (95% CI 0.25 to 8.51), P = 0.68

Nervous

Gornitsky 2004

0/28

1/28

Random‐effects meta‐analysis of 2 studies: RR 1.02 (95% CI 0.11 to 9.33), P = 0.99

Heterogeneity: I2 = 0%, P = 0.33

Lozada‐Nur 1998

1/12

0/12

Rhinitis

Fisher 2003

2/118

5/114

Random‐effects meta‐analysis of 3 studies: RR 0.87 (95% CI 0.41 to 1.86), P = 0.72

Heterogeneity: I2 = 0%, P = 0.53

Lozada‐Nur 1998

1/12

1/12

Sangthawan 2001

8/29

6/24

Blurred vision

Lozada‐Nur 1998

1/12

0/12

RR 3.00 (95% CI 0.13 to 67.06), P = 0.49

Urinary frequency

Fisher 2003

7/118

5/114

Random‐effects meta‐analysis of 2 studies: RR 0.87 (95% CI 0.43 to 1.75), P = 0.70

Heterogeneity: I2 = 0%, P = 0.32

Sangthawan 2001

6/25

8/22

Dizziness

Gornitsky 2004

0/28

2/28

Random‐effects meta‐analysis of 2 studies: RR 0.80 (95% CI 0.18 to 3.45), P = 0.76

Heterogeneity: I2 = 13%, P = 0.28

Sangthawan 2001

4/25

3/22

Palpitation

Sangthawan 2001

0/25

4/22

RR 0.10 (95% CI 0.01 to 1.73), P = 0.11

Skin flushing

Fisher 2003

1/118

0/114

RR 2.90 (95% CI 0.12 to 70.44), P = 0.51

Motor tremors

Fisher 2003

2/118

1/114

RR 1.93 (95% CI 0.18 to 21.02), P = 0.59

Sleep problems

Gornitsky 2004

End of radiotherapy

Mean 37.3 (SD 36.4)

n = 17

Mean 49.6 (SD 36.9)

n = 19

Mean difference ‐12.30 (95% CI ‐36.27 to 11.67), P = 0.31

RTOG (grade 3; mucous membrane, pharynx and larynx)

Warde 2002

No statistically significant difference between treatment groups

CI = confidence interval; RR = risk ratio; RTOG = Radiation Therapy Oncology Group; SD = standard deviation.

Figuras y tablas -
Table 1. Pilocarpine versus no treatment/placebo (other outcomes)
Table 2. Biperiden plus pilocarpine versus no treatment/placebo (other outcomes)

Outcome

Study ID

Time point

Pilocarpine

Control

Results

Comments

Dysphagia (WHO grade 3+)

Rode 1999

12 months after RT

1/30

4/30

RR 0.25 (95% CI 0.03 to 2.11), P = 0.20

CI = confidence interval; RR = risk ratio; RT = radiotherapy; WHO = World Health Organization.

Figuras y tablas -
Table 2. Biperiden plus pilocarpine versus no treatment/placebo (other outcomes)
Table 3. Amifostine versus no treatment/placebo (other outcomes)

Outcome

Study ID

Time point

Amifostine

Control

Results

Comments

Quality of life

Jellema 2006

Assessed at end of RT and 6, 12, 18 and 24 months after RT

No data

No data

"No significant differences between the 3 treatment arms"

Dysphagia (difficulty in swallowing) (0‐4 scale): grade 3 and above

Antonadou 2002

End of RT

14/22

23/23

Random‐effects meta‐analysis of 2 studies: RR 0.50 (95% CI 0.17 to 1.48); P = 0.21

Heterogeneity: I2 = 40%, P = 0.20

Büntzel 1998

1/14

5/14

Antonadou 2002

4 weeks after RT

2/22

3/23

RR 0.70 (95% CI 0.13 to 3.78); P = 0.68

By 8 weeks after RT, no participants had grade 3 or above dysphagia

Dysgeusia (taste disturbance) (0‐4 scale): grade 2 and above

Büntzel 1998

End of RT

3/14

14/14

RR 0.24 (95% CI 0.10 to 0.61); P = 0.003

Cost data (mean per patient supportive care costs)

Büntzel 1998

End of RT

USD 4401

USD 5873

P = 0.02

Vomiting

Antonadou 2002

1/22

0/23

Random‐effects meta‐analysis of 5 studies: RR 4.90 (95% CI 2.87 to 8.38); P < 0.00001

Heterogeneity: I2 = 0%, P = 0.96

Brizel 2000

55/150

11/153

Buentzel 2006

8/66

2/64

He 2004

1/17

0/15

"1 patient left due to gastrointestinal tract reaction/side effect, all other patients completed the treatment"

"At the beginning of treatment, nausea and vomiting was obvious for amifostine group, but after treating with metoclopramide, there was no significant difference between 2 groups in gastrointestinal tract reaction/side effect"

Jellema 2006

10/60

0/31

Peng 2006

10/18

Data not reported in control group. Unknown if this was due to 0 events

Veerasarn 2006

18/32

Data not reported in control group. Unknown if this was due to 0 events

Hypotension

Antonadou 2002

3/22

0/23

Random‐effects meta‐analysis of 3 studies: RR 9.20 (95% CI 2.84 to 29.83); P = 0.0002

Heterogeneity: I2 = 0%, P = 0.88

Brizel 2000

22/150

2/153

Büntzel 1998

2/14

0/14

Veerasarn 2006

5/32

Data not reported in control group. Unknown if this was due to 0 events

Nausea

Brizel 2000

66/150

25/153

Random‐effects meta‐analysis of 4 studies: RR 2.60 (95% CI 1.81 to 3.74); P < 0.00001

Heterogeneity: I2 = 0%, P = 0.45

Buentzel 2006

4/66

4/64

He 2004

1/17

0/15

"1 patient left due to gastrointestinal tract reaction/side effect, all other patients completed the treatment"

"At the beginning of treatment, nausea and vomiting was obvious for amifostine group, but after treating with metoclopramide, there was no significant difference between 2 groups in gastrointestinal tract reaction/side effect"

Jellema 2006

23/60

3/31

Peng 2006

10/18

Data not reported in control group. Unknown if this was due to 0 events

Veerasarn 2006

20/32

Data not reported in control group. Unknown if this was due to 0 events

Allergic response

Brizel 2000

8/150

0/153

Random‐effects meta‐analysis of 3 studies: RR 7.51 (95% CI 1.40 to 40.39); P = 0.02

Heterogeneity: I2 = 0%, P = 0.77

Buentzel 2006

2/66

0/64

Jellema 2006

4/60

0/31

Asthenia (weakness or lack of energy)

Buentzel 2006

3/66

1/64

RR 2.91 (95% CI 0.31 to 27.24); P = 0.35

Alopecia

Vacha 2003

Similar in both groups and increased continuously during the treatment

Skin toxicity

Vacha 2003

Similar in both groups and increased continuously during the treatment

Hot flush

Peng 2006

"..dizziness, fatigue, hiccup, sneezing, facial flush all in less than 5% of the patients"

Veerasarn 2006

17/32

Data not reported in control group. Unknown if this was due to 0 events

Somnolence (drowsiness)

Veerasarn 2006

18/32

Data not reported in control group. Unknown if this was due to 0 events

Sneezing

Peng 2006

"..dizziness, fatigue, hiccup, sneezing, facial flush all in less than 5% of the patients"

Veerasarn 2006

13/32

Data not reported in control group. Unknown if this was due to 0 events

Hiccup

Peng 2006

"..dizziness, fatigue, hiccup, sneezing, facial flush all in less than 5% of the patients"

Veerasarn 2006

10/32

Data not reported in control group. Unknown if this was due to 0 events

Dizziness

Peng 2006

"...dizziness, fatigue, hiccup, sneezing, facial flush all in less than 5% of the patients"

Fatigue

Peng 2006

"..dizziness, fatigue, hiccup, sneezing, facial flush all in less than 5% of the patients"

CI = confidence interval; RR = risk ratio; RT = radiotherapy; USD = US dollars.

Figuras y tablas -
Table 3. Amifostine versus no treatment/placebo (other outcomes)
Table 4. Amifostine: comparison of different doses (other outcomes)

Outcome

Study ID

Time point

Amifostine 3 times weekly

Amifostine 5 times weekly

Results

Comments

Quality of life

Jellema 2006

Assessed at end of RT and 6, 12, 18 and 24 months after RT

No data

No data

"No significant differences between the 3 treatment arms"

Nausea

Jellema 2006

9/30

14/30

RR 0.64 (95% CI 0.33 to 1.25); P = 0.19

Vomiting

Jellema 2006

2/30

8/30

RR 0.25 (95% CI 0.06 to 1.08); P = 0.06

Allergic response

Jellema 2006

2/30

2/30

RR 1.00 (95% CI 0.15 to 6.64); P = 1

CI = confidence interval; RR = risk ratio; RT = radiotherapy.

Figuras y tablas -
Table 4. Amifostine: comparison of different doses (other outcomes)
Table 5. Amifostine: different routes of administration (other outcomes)

Outcome

Study ID

Time point

Intravenous

Subcutaneous

Results

Comments

Nausea/vomiting

Bardet 2011

29%

36%

P = 0.267

Hypotension

Bardet 2011

20%

8%

P = 0.007

Skin rash

Bardet 2011

10%

22%

P = 0.012

Local pain at injection site

Bardet 2011

0%

8%

P = 0.001

Fever

Bardet 2011

2%

0%

P = 0.256

Asthenia (weakness or lack of energy)

Bardet 2011

1%

6%

P = 0.054

Figuras y tablas -
Table 5. Amifostine: different routes of administration (other outcomes)
Table 6. Chinese medicine (other outcomes)

Outcome

Study ID

Intervention

Time point

Study

Control

Results

Comments

Dysphasia (difficulty in swallowing)

(score for EORTC‐H&N35 questionnaire: mean (SD))

Lin 2014

Chinese medicine (TWBXM)

End of RT

50.2 (26.3); n = 35

38.9 (25.9); n = 33

P = 0.07

Lin 2014

Chinese medicine (TWBXM)

1 month after RT

30.2 (29.8); n = 23

26.7 (24.8); n = 21

P = 0.65

Dysgeusia (taste disturbance) (0 to 3 scale): grade 1 and above

Lin 2014

Chinese medicine (TWBXM)

End of RT

32/38

29/33

RR 0.96 (95% CI 0.79 to 1.16); P = 0.13

Speech difficulty

(mean (SD) score for EORTC‐H&N35 questionnaire)

Lin

2014

Chinese medicine (TWBXM)

End of RT

36.3 (26.7); n = 35

28.6 (26.2); n = 33

P = 0.23

Lin

2014

Chinese medicine (TWBXM)

1 month after RT

27.4 (28.6); n = 23

22.7 (19.5); n = 21

P = 0.50

Difficulty in mouth opening

(mean (SD) score for EORTC‐H&N35 questionnaire)

Lin

2014

Chinese medicine (TWBXM)

End of RT

39.6 (28.2); n = 35

41.4 (27.7); n = 33

P = 0.79

Lin

2014

Chinese medicine (TWBXM)

1 month after RT

32.2 (31.5); n = 23

33.3 (24.1); n = 21

P = 0.88

Difficulty in mouth opening

(0 to 2 scale): grade 1 and above

Hu 2005

Chinese medicine (Shenqi Fanghou recipe)

"During the treatment"

22/70

52/70

RR 0.42 (95% CI 0.29 to 0.61); P < 0.001

Skin toxicity (0 to 3 scale): grade 1 and above

Lin

2014

Chinese medicine (TWBXM)

End of RT

35/38

30/33

RR 1.01 (95% CI 0.88 to 1.17); P = 0.82

Skin toxicity (0 to 4 scale): grade 1 and above

Hu 2005

Chinese medicine (Shenqi Fanghou recipe)

"During the treatment"

57/70

68/70

RR 0.84 (95% CI 0.74 to 0.94); P = 0.002

Skin toxicity (prevalence according to RTOG standards)

Han 2010

Chinese medicine (Jinlong capsule)

46.82%

58.32%

Quote: "toxicities during and after treatment were assessed"

Comment: time point for assessment unclear; minor discrepancies in presented data

Nausea/vomiting (0 to 3 scale): grade 1 and above

Lin

2014

Chinese medicine (TWBXM)

End of RT

12/38

4/33

RR 2.61 (95% CI 0.93 to 7.30); P = 0.183

Hoarseness

Lin

2014

Chinese medicine (TWBXM)

End of RT

1/38

3/33

RR 0.29 (95% CI 0.03 to 2.65); P = 0.26

Fatigue

(mean (SD) score for EORTC‐C30 questionnaire)

Lin

2014

Chinese medicine (TWBXM)

End of RT

43.2 (26.2); n = 35

42.4 (23.0); n = 33

P = 0.88

Lin

2014

Chinese medicine (TWBXM)

1 month after RT

31.2 (28.3); n = 23

36.4 (25.0); n = 21

P = 0.51

Pain

(mean (SD) score for EORTC‐C30 questionnaire)

Lin

2014

Chinese medicine (TWBXM)

End of RT

46.8 (23.2); n = 35

41.7 (27.4); n = 33

P = 0.40

Lin

2014

Chinese medicine (TWBXM)

1 month after RT

35.9 (27.0); n = 23

40.9 (29.9); n = 21

P = 0.54

Pain

(mean (SD) score for EORTC‐H&N35 questionnaire)

Lin

2014

Chinese medicine (TWBXM)

End of RT

55.4 (25.1); n = 35

42.4 (20.5); n = 33

P = 0.02

Lin

2014

Chinese medicine (TWBXM)

1 month after RT

31.6 (24.2); n = 23

37.8 (23.3); n = 21

P = 0.35

Dyspnea

(mean (SD) score for EORTC‐C30 questionnaire)

Lin

2014

Chinese medicine (TWBXM)

End of RT

17.1 (23.1); n = 35

16.7 (20.7); n = 33

P = 0.93

Lin

2014

Chinese medicine (TWBXM)

1 month after RT

20.5 (21.2); n = 23

13.6 (22.2); n = 21

P = 0.28

Insomnia

(mean (SD) score for EORTC‐C30 questionnaire)

Lin

2014

Chinese medicine (TWBXM)

End of RT

40.5 (25.0); n = 35

31.2 (25.3); n = 33

P = 0.13

Lin

2014

Chinese medicine (TWBXM)

1 month after RT

30.8 (24.8); n = 23

31.8 (28.1); n = 21

P = 0.28

Appetite loss

(mean (SD) score for EORTC‐C30 questionnaire)

Lin

2014

Chinese medicine (TWBXM)

End of RT

45.0 (30.7); n = 35

45.8 (29.0); n = 33

P = 0.91

Lin

2014

Chinese medicine (TWBXM)

1 month after RT

28.2 (26.1); n = 23

34.9 (30.0); n = 21

P = 0.42

Constipation

(mean (SD) score for EORTC‐C30 questionnaire)

Lin

2014

Chinese medicine (TWBXM)

End of RT

37.8 (27.4); n = 35

29.2 (20.3); n = 33

P = 0.15

Lin

2014

Chinese medicine (TWBXM)

1 month after RT

29.5 (30.3); n = 23

25.7 (20.4); n = 21

P = 0.63

Diarrhoea

(mean (SD) score for EORTC‐C30 questionnaire)

Lin

2014

Chinese medicine (TWBXM)

End of RT

9.0 (15.0); n = 35

6.2 (13.2); n = 33

P = 0.42

Lin

2014

Chinese medicine (TWBXM)

1 month after RT

9.0 (15.1); n = 23

6.1 (16.7); n = 21

P = 0.53

Adverse effects

Han 2010

Chinese medicine (Jinlong capsule)

Leukopenia, nausea, vomiting, 1 participant had dizziness and blood pressure drop, 1 participant had skin rash

Not reported

Adverse effects

Hu 2005

Chinese medicine (Shenqi Fanghou recipe)

"During the treatment"

No adverse event

Not reported

CI = confidence interval; EORTC = European Organisation for Research and Treatment of Cancer; H&N = head and neck; RR = risk ratio; RT = radiotherapy; RTOG = Radiation Therapy Oncology Group; SD = standard deviation; TWBXM = Tianwang Buxin Mini‐pills.

Figuras y tablas -
Table 6. Chinese medicine (other outcomes)
Table 7. Palifermin versus placebo (other outcomes)

Outcome

Study ID

Time point

Palifermin

Placebo

Results

Oral related symptoms (other than salivary gland dysfunction/xerostomia)

Dysphagia

Le 2011

3 months postRT

29/94

19/91

Random‐effects meta‐analysis of 3 studies: RR 1.32 (95% CI 0.55 to 3.13); P = 0.54

Heterogeneity: I2 = 94%, P < 0.00001

Brizel 2008

61/64

31/32

Henke 2011

32/92

20/93

Mouth and throat soreness ‐ 0 (no soreness) to 4 (extreme soreness) OMWQ‐HN scale

Le 2011

3 months postRT

n = 94, mean = 1.66, SD = 0.73

n = 94, mean = 1.86, SD = 0.65

Random‐effects meta‐analysis of 2 studies: mean difference ‐0.12 (95% CI ‐0.27 to 0.02); P = 0.10

Heterogeneity: I2 = 13%, P = 0.28

Henke 2011

n = 92, mean = 1.52, SD = 0.69

n = 94, mean = 1.57, SD = 0.63

Adverse events

Nausea

Le 2011

47/94

42/91

Random‐effects meta‐analysis of 2 studies: RR 0.96 (95% CI 0.77 to 1.19); P = 0.69

Heterogeneity: I2 = 28%, P = 0.24

Brizel 2008

48/67

26/32

Fever

Brizel 2008

30/67

13/32

RR 1.10 (95% CI 0.67 to 1.81); P = 0.70

Constipation

Le 2011

31/94

24/91

Random‐effects meta‐analysis of 2 studies: RR 1.15 (95% CI 0.82 to 1.60); P = 0.42

Heterogeneity: I2 = 0%, P = 0.57

Brizel 2008

28/67

13/32

Diarrhoea

Brizel 2008

14/67

8/32

Random‐effects meta‐analysis of 2 studies: RR 1.28 (95% CI 0.49 to 3.36); P = 0.61

Heterogeneity: I2 = 57%, P = 0.13

Henke 2011

11/92

5/93

Insomnia

Brizel 2008

12/67

4/32

Random‐effects meta‐analysis of 2 studies: RR 0.77 (95% CI 0.23 to 2.55); P = 0.67

Heterogeneity: I2 = 63%, P = 0.10

Henke 2011

5/92

12/93

Dyspnea

Brizel 2008

9/67

1/32

RR 1.10 (95% CI 0.67 to 1.81); P = 0.70

Cough

Brizel 2008

8/67

5/32

RR 0.76 (95% CI 0.27 to 2.15); P = 0.61

Headache

Brizel 2008

8/67

2/32

Random‐effects meta‐analysis of 2 studies: RR 2.13 (95% CI 0.86 to 5.28); P = 0.10

Heterogeneity: I2 = 0%, P = 0.86

Henke 2011

9/92

4/93

Decreased weight

Le 2011

29/94

27/91

Random‐effects meta‐analysis of 2 studies: RR 1.01 (95% CI 0.67 to 1.52); P = 0.96

Heterogeneity: I2 = 0%, P = 0.73

Brizel 2008

7/67

4/32

Dizziness

Brizel 2008

5/67

4/32

RR 0.60 (95% CI 0.17 to 2.07); P = 0.42

Anxiety

Brizel 2008

4/67

5/32

RR 0.38 (95% CI 0.11 to 1.33); P = 0.13

Hypomagnesemia

Brizel 2008

4/67

4/32

RR 0.48 (95% CI 0.13 to 1.79); P = 0.27

Vomiting

Le 2011

26/94

26/91

Random‐effects meta‐analysis of 2 studies: RR 0.98 (95% CI 0.72 to 1.33); P = 0.89

Heterogeneity: I2 = 0%, P = 0.96

Brizel 2008

33/67

16/32

Radiation skin injury

Le 2011

25/94

13/91

RR 1.10 (95%CI 0.67 to 1.81); P = 0.70

Anaemia

Le 2011

21/94

34/91

Random‐effects meta‐analysis of 2 studies: RR 0.83 (95% CI 0.33 to 2.05); P = 0.68

Heterogeneity: I2 = 54%, P = 0.14

Brizel 2008

10/67

3/32

Fatigue

Le 2011

21/94

20/91

Random‐effects meta‐analysis of 3 studies: RR 0.88 (95% CI 0.60 to 1.30); P = 0.52

Heterogeneity: I2 = 2%, P = 0.36

Henke 2011

7/92

14/93

Brizel 2008

17/67

8/32

Leukopenia

Le 2011

21/94

12/91

Random‐effects meta‐analysis of 2 studies: RR 1.01 (95% CI 0.37 to 2.78); P = 0.98

Heterogeneity: I2 = 79%, P = 0.03

Henke 2011

12/92

20/93

Granulocytopenia

Brizel 2008

20/67

6/32

RR 1.59 (95% CI 0.47 to 5.39); P = 0.45

Pharyngolaryngeal pain

Le 2011

20/94

23/91

RR 0.84 (95% CI 0.50 to 1.42); P = 0.52

Hypokalemia

Le 2011

19/94

8/91

RR 2.04 (95% CI 0.98 to 4.28); P = 0.06

Pyrexia

Le 2011

16/94

19/91

RR 0.82 (95% CI 0.45 to 1.48); P = 0.50

Mucosal inflammation

Henke 2011

4/92

10/93

RR 0.40 (95% CI 0.13 to 1.24); P = 0.11

Asthenia

Henke 2011

13/92

7/93

RR 1.88 (95% CI 0.78 to 4.49); P = 0.16

Abdominal pain

Henke 2011

7/92

2/93

RR 3.54 (95% CI 0.75 to 16.58); P = 0.11

Back pain

Henke 2011

6/92

1/93

RR 6.07 (95% CI 0.74 to 49.40); P = 0.09

Febrile neutropenia

Henke 2011

1/92

Considered "serious adverse event"

0/93

RR 3.03 (95% CI 0.13 to 73.48); P = 0.50

Dehydration

Le 2011

13/94

19/91

Random‐effects meta‐analysis of 3 studies: RR 0.75 (95% CI 0.45 to 1.25); P = 0.27

Heterogeneity: I2 = 30%, P = 0.24

Henke 2011

6/92

13/93

Brizel 2008

20/67

8/32

CI = confidence interval; OMWQ‐HN = Oral Mucositis Weekly Questionnaire ‐ Head and Neck Cancer; RR = risk ratio; RT = radiotherapy; SD = standard deviation.

Figuras y tablas -
Table 7. Palifermin versus placebo (other outcomes)
Table 8. Bethanechol versus placebo (other outcomes)

Outcome

Study ID

Results

Adverse effects

Jaguar 2015

No statistical difference between the groups in bethanechol‐related toxicity. Quote: "No patient experienced severe (grade 3) toxicity and no one dropped out of the study due to adverse effects"

Figuras y tablas -
Table 8. Bethanechol versus placebo (other outcomes)
Table 9. Bethanechol versus artificial saliva (other outcomes)

Outcome

Study ID

Time point

Bethanechol

Artificial saliva

Results

Comments

Lacrimation (watering eyes)

Jham 2007

End of RT

3/22

0/21

RR 6.70 (95% CI 0.37 to 122.29); P = 0.2

Nervousness

3/22

0/21

RR 6.70 (95% CI 0.37 to 122.29); P = 0.2

Frequent urination

3/22

0/21

RR 6.70 (95% CI 0.37 to 122.29); P = 0.2

Sweating

2/22

0/21

RR 4.78 (95% CI 0.24 to 94.12); P = 0.3

"1 patient using bethanechol dropped out of the study due to excessive sweating (Grade 2 severity; National Cancer Institute Common Terminology Criteria for Adverse Events – NCI CTCAE, v 3"

Warm face

2/22

0/21

RR 4.78 (95% CI 0.24 to 94.12); P = 0.3

Cramps

1/22

0/21

RR 2.87 (95% CI 0.12 to 66.75); P = 0.51

Diarrhoea

1/22

0/21

RR 2.87 (95% CI 0.12 to 66.75); P = 0.51

Nausea

1/22

2/21

RR 0.48 (95% CI 0.05 to 4.88); P = 0.53

CI = confidence interval; RR = risk ratio; RT = radiotherapy.

Figuras y tablas -
Table 9. Bethanechol versus artificial saliva (other outcomes)
Table 10. Selenium versus no intervention (other outcomes)

Outcome

Study ID

Time point

Reported in text

Loss of taste

Büntzel 2010

6 weeks after end RT

"Ageusia was milder in the selenium group. But the difference was not significant"

Dysphagia

Büntzel 2010

6 weeks after end RT

"The only significant difference was observed at week 7, when the selenium group had developed a mean value of 1.533 versus 2.167 in the control group (P = 0.05)"

Adverse events

Büntzel 2010

6 weeks after end RT

"23 serious adverse events (SAEs) were seen in the selenium group, compared to 22 in the control group (P = 0.476). No statistically significant differences in toxicities were found using the 2‐tailed Fisher's exact test"

RT = radiotherapy.

Figuras y tablas -
Table 10. Selenium versus no intervention (other outcomes)
Table 11. Antiseptic mouthrinse versus placebo (other outcomes)

Outcome

Study ID

Time point

Antiseptic rinse

Placebo

Results

Drooling

Lanzós 2010

4 weeks from baseline

Increased 6

No change or decreased 8

6/14

Increased 3

No change or decreased 7

3/10

RR 1.43 (95% CI 0.46 to 4.39); P = 0.53

Adverse events

Lanzós 2010

"No relevant adverse events were reported in any group"

CI = confidence interval; RR = risk ratio.

Figuras y tablas -
Table 11. Antiseptic mouthrinse versus placebo (other outcomes)
Table 12. Antimicrobial lozenge versus placebo (other outcomes)

Outcome

Study ID

Time point

Antimicrobial lozenge

Placebo

Results

Mouth pain

Duncan 2005

Worse over 6 months

32/66

32/62

RR 0.94 (95% CI 0.66 to 1.33); P=0.72

Sore/burning mouth

Duncan 2005

Worse over 6 months

32/65

32/62

RR 0.95 (95% CI 0.68 to 1.35); P=0.79

Throat pain

Duncan 2005

Worse over 6 months

29/66

36/65

RR 0.79 (95% CI 0.56 to 1.12); P=0.19

Dryness in mouth

Duncan 2005

Worse over 6 months

55/66

46/65

RR 1.18 (95% CI 0.97 to 1.42); P=0.09

Nausea

Duncan 2005

Worse over 6 months

27/66

14/65

RR 1.90 (95% CI 1.10 to 3.28); P=0.02

Diarrhoea

Duncan 2005

Worse over 6 months

6/66

3/65

RR 1.97 (95% CI 0.51 to 7.54); P=0.32

Constipation

Duncan 2005

Worse over 6 months

24/66

26/65

RR 0.91 (95% CI 0.59 to 1.42); P=0.67

CI = confidence interval; RR = risk ratio.

Figuras y tablas -
Table 12. Antimicrobial lozenge versus placebo (other outcomes)
Table 13. Polaprezinc versus azulene rinse (other outcomes)

Outcome

Study ID

Time point

Polaprezinc

Azulene rinse

Results

Pain > 2 (0‐3 scale)

Watanabe 2010

Over RT period

5/16

13/15

RR 0.36 (95% CI 0.17 to 0.77); P = 0.008

Taste disturbance > 2 (0‐3 scale)

Watanabe 2010

Over RT period

1/16

8/15

RR 0.12 (95% CI 0.02 to 0.83); P = 0.03

Disability of oral intake

Watanabe 2010

Over RT period

2/16

6/15

RR 0.31 (95% CI 0.07 to 1.31); P = 0.11

CI = confidence interval; RR = risk ratio; RT = radiotherapy.

Figuras y tablas -
Table 13. Polaprezinc versus azulene rinse (other outcomes)
Table 14. Venalot Depot (coumarin/troxerutin) versus placebo

Outcome

Study ID

Results

Adverse events

Grötz 2001

"No adverse events could be attributed to the experimental medication"

Figuras y tablas -
Table 14. Venalot Depot (coumarin/troxerutin) versus placebo
Comparison 1. Pilocarpine versus no treatment/placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia Show forest plot

6

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

1.1 End of radiotherapy

4

122

Std. Mean Difference (IV, Random, 95% CI)

0.20 [‐0.16, 0.56]

1.2 Up to and including 3 months postradiotherapy

3

125

Std. Mean Difference (IV, Random, 95% CI)

0.02 [‐0.33, 0.37]

1.3 Up to and including 6 months postradiotherapy

2

126

Std. Mean Difference (IV, Random, 95% CI)

‐0.35 [‐1.04, 0.33]

2 Xerostomia (LENT‐SOMA scale) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.1 Up to and including 6 months postradiotherapy

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

3 Xerostomia Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3.1 End of radiotherapy

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.2 Up to and including 3 months postradiotherapy

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Salivary flow rate (unstimulated) Show forest plot

3

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 End of radiotherapy

3

76

Std. Mean Difference (IV, Random, 95% CI)

0.24 [‐0.24, 0.72]

4.2 Up to and including 3 months postradiotherapy

1

24

Std. Mean Difference (IV, Random, 95% CI)

‐0.27 [‐1.07, 0.54]

5 Salivary flow rate (stimulated) Show forest plot

3

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

5.1 End of radiotherapy

2

58

Std. Mean Difference (IV, Random, 95% CI)

0.08 [‐0.44, 0.59]

5.2 Up to and including 3 months postradiotherapy

1

24

Std. Mean Difference (IV, Random, 95% CI)

0.40 [‐0.41, 1.21]

5.3 Up to and including 6 months postradiotherapy

1

9

Std. Mean Difference (IV, Random, 95% CI)

0.52 [‐0.84, 1.87]

5.4 Up to and including 12 months postradiotherapy

1

9

Std. Mean Difference (IV, Random, 95% CI)

0.53 [‐0.83, 1.88]

6 Salivary flow rate (> 0 g) unstimulated Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

6.1 End of radiotherapy

1

154

Risk Ratio (M‐H, Random, 95% CI)

1.90 [0.98, 3.69]

6.2 Up to and including 3 months postradiotherapy

1

152

Risk Ratio (M‐H, Random, 95% CI)

2.0 [0.86, 4.68]

7 Salivary flow rate (> 0 g) stimulated Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

7.1 End of radiotherapy

1

138

Risk Ratio (M‐H, Random, 95% CI)

1.87 [0.77, 4.52]

7.2 Up to and including 3 months postradiotherapy

1

139

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.23, 2.11]

8 Overall survival Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

9 Quality of life Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Totals not selected

9.1 End of radiotherapy

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

9.2 Up to and including 3 months postradiotherapy

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

9.3 Up to and including 6 months postradiotherapy

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 1. Pilocarpine versus no treatment/placebo
Comparison 2. Biperiden plus pilocarpine versus no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Salivary flow rate (unstimulated) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

1.1 End of radiotherapy

1

40

Mean Difference (IV, Random, 95% CI)

0.02 [‐0.08, 0.12]

1.2 Up to and including 3 months postradiotherapy

1

17

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

2 Salivary flow rate (> 0 g) unstimulated Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 End of radiotherapy

1

60

Risk Ratio (M‐H, Random, 95% CI)

1.11 [0.77, 1.58]

2.2 Up to and including 3 months postradiotherapy

1

60

Risk Ratio (M‐H, Random, 95% CI)

7.50 [1.88, 29.99]

2.3 Up to and including 6 months postradiotherapy

1

60

Risk Ratio (M‐H, Random, 95% CI)

29.00 [1.81, 465.07]

2.4 Up to and including 12 months postradiotherapy

1

60

Risk Ratio (M‐H, Random, 95% CI)

35.00 [2.20, 556.71]

Figuras y tablas -
Comparison 2. Biperiden plus pilocarpine versus no treatment
Comparison 3. Amifostine versus no treatment/placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (0 to 4 scale ‐ grade 2 or above) Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 End of radiotherapy

3

119

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.19, 0.67]

1.2 Up to and including 3 months postradiotherapy

5

687

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.48, 0.92]

1.3 12 months postradiotherapy

7

682

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.40, 1.23]

2 Salivary flow rate (unstimulated) Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

2.1 End of radiotherapy

2

83

Mean Difference (IV, Random, 95% CI)

0.34 [0.07, 0.61]

2.2 Up to and including 3 months postradiotherapy

1

41

Mean Difference (IV, Random, 95% CI)

0.13 [‐0.90, 1.16]

2.3 12 months postradiotherapy

1

27

Mean Difference (IV, Random, 95% CI)

0.32 [0.09, 0.55]

3 Salivary flow rate (unstimulated) ‐ incidence of > 0.1 g in 5 min Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 12 months postradiotherapy

1

175

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.13, 1.86]

4 Salivary flow rate (stimulated) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 End of radiotherapy

1

47

Mean Difference (IV, Random, 95% CI)

‐0.09 [‐1.48, 1.30]

4.2 Up to and including 3 months postradiotherapy

1

41

Mean Difference (IV, Random, 95% CI)

0.38 [‐1.43, 2.19]

4.3 12 months postradiotherapy

1

27

Mean Difference (IV, Random, 95% CI)

0.82 [‐0.47, 2.11]

5 Salivary flow rate (stimulated) ‐ incidence of > 0.1 g in 5 min Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 12 months postradiotherapy

1

173

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.89, 1.41]

6 Overall survival at 12 to 24 months postradiotherapy Show forest plot

2

271

Hazard ratio (Random, 95% CI)

1.18 [0.85, 1.66]

7 Overall survival ‐ narrative data Show forest plot

Other data

No numeric data

8 Progression‐free survival at 12 to 24 months postradiotherapy Show forest plot

2

247

Hazard ratio (Random, 95% CI)

0.94 [0.70, 1.27]

9 Progression‐free survival Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

9.1 18 months postradiotherapy

1

45

Risk Ratio (M‐H, Random, 95% CI)

1.11 [0.81, 1.51]

10 Progression‐free survival ‐ narrative data Show forest plot

Other data

No numeric data

11 Locoregional tumour control at 12 to 24 months postradiotherapy Show forest plot

2

279

Hazard ratio (Random, 95% CI)

0.90 [0.74, 1.11]

12 Locoregional tumour control ‐ narrative data Show forest plot

Other data

No numeric data

13 Disease‐free survival Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

13.1 24 months postradiotherapy

1

170

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.73, 1.21]

14 Disease‐free survival Show forest plot

Other data

No numeric data

15 Quality of life (Patient Benefit Questionnaire) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

15.1 End of radiotherapy

1

298

Mean Difference (IV, Random, 95% CI)

0.38 [‐0.07, 0.83]

15.2 Up to and including 3 months postradiotherapy

1

233

Mean Difference (IV, Random, 95% CI)

0.52 [‐0.02, 1.06]

15.3 12 months postradiotherapy

1

180

Mean Difference (IV, Random, 95% CI)

0.70 [0.20, 1.20]

Figuras y tablas -
Comparison 3. Amifostine versus no treatment/placebo
Comparison 4. Amifostine (comparison of dosages)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (0 to 4 scale ‐ grade 2 or above) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 12 months postradiotherapy

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.58, 1.53]

2 Overall survival ‐ narrative data Show forest plot

Other data

No numeric data

3 Locoregional tumour control ‐ narrative data Show forest plot

Other data

No numeric data

Figuras y tablas -
Comparison 4. Amifostine (comparison of dosages)
Comparison 5. Amifostine (intravenous versus subcutaneous)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (0 to 4 scale ‐ grade 2 or above) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Up to and including 3 months postradiotherapy

1

263

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.76, 1.40]

1.2 12 months postradiotherapy

1

127

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.42, 0.88]

2 Overall survival Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

2.1 48 months after radiotherapy

1

Hazard Ratio (Random, 95% CI)

1.36 [0.89, 2.10]

3 Locoregional tumour control Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

3.1 48 months after radiotherapy

1

Hazard Ratio (Random, 95% CI)

1.34 [0.76, 2.36]

Figuras y tablas -
Comparison 5. Amifostine (intravenous versus subcutaneous)
Comparison 6. Chinese medicine versus no treatment/placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 End of radiotherapy: Shenqi Fanghou recipe versus no intervention

1

140

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.28, 0.55]

1.2 End of radiotherapy: TWBXM versus placebo

1

71

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.78, 1.03]

1.3 Up to and including 3 months postradiotherapy: Jinlong capsules versus no intervention

1

95

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.59, 1.36]

2 Xerostomia Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

2.1 End of radiotherapy: TWBXM versus placebo

1

68

Mean Difference (IV, Random, 95% CI)

‐2.41 [‐16.19, 11.37]

2.2 Up to and including 3 months postradiotherapy: TWBXM versus placebo

1

44

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐17.21, 17.01]

3 Salivary flow rate (stimulated) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 End of radiotherapy: Chinese medicine versus no intervention

1

50

Mean Difference (IV, Random, 95% CI)

0.09 [0.03, 0.15]

4 Overall survival (12 months postRT) Show forest plot

1

78

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.84, 1.30]

5 Quality of life (EORTC‐C30) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

5.1 End of radiotherapy: TWBXM versus placebo

1

68

Mean Difference (IV, Random, 95% CI)

2.39 [‐8.74, 13.52]

5.2 Up to and including 3 months postradiotherapy: TWBXM versus placebo

1

44

Mean Difference (IV, Random, 95% CI)

1.93 [‐13.04, 16.90]

Figuras y tablas -
Comparison 6. Chinese medicine versus no treatment/placebo
Comparison 7. Palifermin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (0 to 4 scale ‐ grade 2 or above) Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Up to and including 3 months postRT

3

471

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.77, 1.22]

2 Overall survival at 42 to 72 months from baseline Show forest plot

3

Hazard Ratio (Random, 95% CI)

1.00 [0.72, 1.39]

3 Progression‐free survival at 42 to 72 months from baseline Show forest plot

3

Hazard Ratio (Random, 95% CI)

1.06 [0.80, 1.42]

Figuras y tablas -
Comparison 7. Palifermin versus placebo
Comparison 8. Bethanechol versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (0 to 3 scale ‐ grade 2 or above) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 End of radiotherapy

1

84

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.28, 0.66]

1.2 Up to and including 3 months postradiotherapy

1

84

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.65, 1.01]

2 Salivary flow rate (unstimulated) ‐ ml/min Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

2.1 2 months postradiotherapy

1

97

Mean Difference (IV, Random, 95% CI)

0.19 [0.06, 0.32]

3 Salivary flow rate (stimulated) ‐ ml/min Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 2 months postradiotherapy

1

97

Mean Difference (IV, Random, 95% CI)

0.15 [‐0.03, 0.33]

Figuras y tablas -
Comparison 8. Bethanechol versus placebo
Comparison 9. Bethanechol versus artificial saliva

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (dry mouth? yes/no) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 End of radiotherapy

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.30, 1.29]

1.2 8 to 40 weeks postradiotherapy

1

30

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.30, 1.05]

2 Salivary flow rate (unstimulated) ‐ ml/min Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

2.1 End of radiotherapy

1

36

Mean Difference (IV, Random, 95% CI)

0.12 [0.01, 0.23]

2.2 8 to 40 weeks postradiotherapy

1

33

Mean Difference (IV, Random, 95% CI)

0.07 [‐0.02, 0.16]

3 Salivary flow rate (stimulated) ‐ ml/min Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 End of radiotherapy

1

32

Mean Difference (IV, Random, 95% CI)

0.13 [‐0.03, 0.29]

3.2 8 to 40 weeks postradiotherapy

1

29

Mean Difference (IV, Random, 95% CI)

0.21 [0.01, 0.41]

4 Overall survival Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 40 weeks postradiotherapy

1

43

Risk Ratio (M‐H, Random, 95% CI)

1.59 [0.43, 5.84]

Figuras y tablas -
Comparison 9. Bethanechol versus artificial saliva
Comparison 10. Selenium versus no selenium

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia Show forest plot

Other data

No numeric data

Figuras y tablas -
Comparison 10. Selenium versus no selenium
Comparison 11. Antimicrobial lozenge versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (QoL response for dryness) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Up to and including 3 months postradiotherapy

1

133

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.97, 1.40]

2 Quality of life Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Up to and including 3 months postradiotherapy (change score over 6 months)

1

131

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.65, 1.50]

Figuras y tablas -
Comparison 11. Antimicrobial lozenge versus placebo
Comparison 12. Polaprezinc versus azulene oral rinse

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Xerostomia (grade 2 or above) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 End of radiotherapy

1

31

Risk Ratio (M‐H, Random, 95% CI)

0.17 [0.04, 0.65]

Figuras y tablas -
Comparison 12. Polaprezinc versus azulene oral rinse