Scolaris Content Display Scolaris Content Display

Duloxetina para el tratamiento de la neuropatía dolorosa, el dolor crónico o la fibromialgia

Contraer todo Desplegar todo

Referencias

Arnold 2004 {published data only}

Arnold LM, Lu Y, Crofford LJ, Wohlreich M, Detke MJ, Iyengar S, et al for the Duloxetine Fibromyalgia Trial Group. A double‐blind multicenter trial comparing duloxetine with placebo in the treatment of fibromyalgia patients with or without major depressive disorder. Arthritis and Rheumatism 2004;50(9):2974‐84. [PUBMED: 15457467]

Arnold 2005 {published data only}

Arnold LM, Rosen A, Pritchett YL, D'Souza DN, Goldstein DJ, Iyengar S, et al. A randomized double‐blind, placebo‐controlled trial of duloxetine in the treatment of women with fibromyalgia with or without major depressive disorder. Pain 2005;119(1‐3):5‐15. [PUBMED: 16298061]

Arnold 2010 {published data only}

Arnold LM, Clauw D, Wang F, Ahl J, Gaynor PJ, Wohlreich MM. Flexible dosed duloxetine in the treatment of fibromyalgia: a randomized double‐blind placebo‐controlled trial [A study comparing duloxetine and placebo in the treatment of fibromyalgia]. Journal of Rheumatology 2010;37(12):2578‐86. [PUBMED: 20843911]

Arnold 2012 {published data only}

Arnold LM, Zhang S, Pangallo BA. Efficacy and safety of duloxetine 30 mg/d in patients with fibromyalgia: a randomized, double‐blind, placebo‐controlled study. Clinical Journal of Pain 2012;28(9):775‐81. [PUBMED: 22971669]

Brecht 2007 {published data only}

Brecht S, Courtecuisse C, Debieuvre C, Croenlein J, Desaiah D, Raskin J, et al. Efficacy and safety of duloxetine 60 mg once daily in the treatment of pain in patients with major depressive disorder and at least moderate pain of unknown etiology: a randomized controlled trial. Journal of Clinical Psychiatry 2007;68(11):1707‐16. [PUBMED: 18052564]

Chappell 2008 {published data only}

Chappell AS, Bradley LA, Wiltse C, Detke MJ, D'Souza DN, Spaeth M. A six‐month double‐blind, placebo‐controlled, randomized clinical trial of duloxetine for the treatment of fibromyalgia. International Journal of General Medicine 2009;30(1):91‐102. [PUBMED: 20428412]

Gao 2010 {published data only}

Gao Y, Ning G, Jia WP, Zhou ZG, Xu ZR, Liu ZM, et al. Duloxetine versus placebo in the treatment of patients with diabetic peripheral neuropathic pain in China. Chinese Medical Journal 2010;123(22):3184‐92. [CTG: NCT00408993; PUBMED: 21163113]

Gaynor 2011a {published data only}

Gaynor PJ, Gopal M, Zheng W, Martinez JM, Robinson MJ, Hann D, Marangell LB. Duloxetine versus placebo in the treatment of major depressive disorder and associated painful physical symptoms: a replication study. Current Medical Research and Opinion 2011;27(10):1859‐67. [PUBMED: 21838410]

Gaynor 2011b {published data only}

Gaynor PJ, Gopal M, Zheng W, Martinez JM, Robinson MJ, Marangell LB. A randomized placebo‐controlled trial of duloxetine in patients with major depressive disorder and associated painful physical symptoms. Current Medical Research and Opinion 2011;27(10):1849‐58. [PUBMED: 21838411]

Goldstein 2005 {published data only}

Goldstein DJ, Lu Y, Detke MJ, Lee TC, Iyengar S. Duloxetine vs. placebo in patients with painful diabetic neuropathy. Pain 2005;116(1‐2):109‐18. [PUBMED: 15927394]

Kaur 2011 {published data only}

Kaur H, Hota D, Bhansali A, Dutta P, Bansal D, Chakrabarti A. A comparative evaluation of amitriptyline and duloxetine in painful diabetic neuropathy: a randomized, double‐blind, cross‐over clinical trial. Diabetes Care 2011;34(4):818‐22. [PUBMED: 21355098]

Raskin 2005 {published data only}

Raskin J, Pritchett YL, Wang F, D'Souza DN, Waninger AL, Iyengar S, et al. A double‐blind, randomized multicentre trial comparing duloxetine with placebo in the management of diabetic peripheral neuropathic pain. Pain Medicine 2005;6(5):346‐56. [PUBMED: 16266355]

Rowbotham 2012 {published data only}

Rowbotham MC, Arslanian A, Nothaft W, Duan WR, Best AE, Pritchett Y, et al. Efficacy and safety of the α4β2 neuronal nicotinic receptor agonist ABT‐894 in patients with diabetic peripheral neuropathic pain [Safety and efficacy study in subjects with diabetic neuropathic pain]. Pain 2012;153(4):862‐8. [CTG: NCT00507936; PUBMED: 22386472]

Russell 2008 {published data only}

Russell IJ, Mease PJ, Smith TR, Kajdasz DK, Wohlreich MM, Detke MJ, et al. Efficacy and safety of duloxetine for the treatment of fibromyalgia in patients with or without major depressive disorder: results from a 6‐month randomized double‐blind, placebo‐controlled, fixed‐dose trial. Pain 2008;136(3):432‐44. [PUBMED: 18395345]

Tesfaye 2013 {published and unpublished data}

Tesfaye S, Wilhelm S, Lledo A, Schacht A, Tölle T, Bouhassira D, et al. Duloxetine and pregabalin: high‐dose monotherapy or their combination? The “COMBO‐DN study” – a multinational, randomized, double‐blind, parallel‐group study in patients with diabetic peripheral neuropathic pain [Use of duloxetine or pregabalin in monotherapy versus combination therapy of both drugs in patients with painful diabetic neuropathy "The COMBO ‐ DN (COmbination vs Monotherapy of pregaBalin and dulOxetine in Diabetic Neuropathy) Study"]. Pain 2013 May 31 [Epub ahead of print]. [CTG: NCT01089556; PUBMED: 23732189]

Vranken 2011 {published data only}

Vranken JH, Hollmann MW, van der Vegt MH, Kruis MR, Heesen M, Vos K, et al. Duloxetine in patients with central neuropathic pain caused by spinal cord injury or stroke: a randomized, double‐blind, placebo‐controlled trial. Pain 2011;152(2):267‐73.

Wernicke 2006 {published data only}

Wernicke JF, Pritchett YL, D'Souza DN, Waninger A, Tran P, Iyengar S, et al. A randomized controlled trial of duloxetine in diabetic peripheral neuropathic pain. Neurology 2006;67(8):1411‐20. [PUBMED: 17060567]

Yasuda 2010 {published and unpublished data}

Yasuda H, Hotta N, Nakao K, Kasuga M, Kashiwagi A, Kawamori R. Superiority of duloxetine to placebo in improving painful diabetic neuropathic pain: results of a randomized controlled trial in Japan [A superiority study of LY248686 versus placebo in the treatment of patients with diabetic peripheral neuropathic pain]. Journal of Diabetes Investigation 2011;2(2):132‐9.

Boyle 2012 {published data only}

Boyle J, Eriksson ME, Gribble L, Gouni R, Johnsen S, Coppini DV, Kerr D. Randomized, placebo‐controlled comparison of amitriptyline, duloxetine, and pregabalin in patients with chronic diabetic peripheral neuropathic pain: impact on pain, polysomnographic sleep, daytime functioning, and quality of life. [Effects of pregabalin, duloxetine & amitriptyline on pain & sleep (NCT00370656)]. Diabetes Care. 2012;35(12):2451‐8. [DOI: 10.2337/dc12‐0656]

Brannan 2005 {published data only}

Brannan SK, Mallinckrodt CH, Brown EB, Wohlreich MM, Watkin JG, Schatzberg AF. Duloxetine 60mg once daily in the treatment of painful physical symptoms in patients with major depressive disorder. Journal of Psychiatric Research 2005;39(1):43‐53. [PUBMED: 15504423]

Canovas 2007 {published data only}

Cánovas L, Illodo G, Castro M, Mouriz L, Vázquez‐Martínez A, Centeno J, et al. Effects of duloxetin and amitriptilin in neuropathic pain: study in 180 patients [Efectos de duloxetina y amitriptilina en el dolor neuropatico: estudio en 180 casos]. Revista de la Sociedad Espanola del Dolor 2007;14(8):568‐73.

Chappell 2009 {published data only}

Chappell AS, Ossanna MJ, Liu‐Seifert H, Iyengar S, Skljarevski V, Li LC, et al. Duloxetine, a centrally acting analgesic, in the treatment of patients with osteoarthritis knee pain: a 13‐week, randomized, placebo‐controlled trial. Pain 2009;146(3):253‐60. [PUBMED: 19625125]

Chappell 2011 {published data only}

Chappell AS, Desaiah D, Liu‐Seifert H, Zhang S, Skljarevski V, Belenkov Y, et al. A double‐blind, randomized, placebo‐controlled study of the efficacy and safety of duloxetine for the treatment of chronic pain due to osteoarthritis of the knee. Pain Practice 2011;11(1):33‐41. [PUBMED: 20602715]

Goldstein 2004 {published data only}

Goldstein DJ, Lu Y, Detke MJ, Wiltse C, Mallinckrodt C, Demitrack MA. Duloxetine in the treatment of depression: a double‐blind placebo‐controlled comparison with paroxetine. Journal of Clinical Psychopharmacology 2004;24(4):389‐99. [PUBMED: 15232330]

Harrison 2013 {published data only}

Harrison T, Miyahara S, Lee A, Evans S, Bastow B, Simpson D, et al. ACTG A5252 Team. Experience and challenges presented by a multicenter crossover study of combination analgesic therapy for the treatment of painful HIV‐associated polyneuropathies [Combination Pain Therapy in HIV Neuropathy]. Pain Medicine 2013;14(7):1039‐47.

Lavoie Smith 2012 {published data only}

Lavoie Smith EM, Pang H, Cirrincione C, Fleishman SB, Paskett ED, Fadul CE, et al. CALGB 170601: A phase III double blind trial of duloxetine to treat painful chemotherapy‐induced peripheral neuropathy (CIPN). Journal of clinical oncology 2012;Conference:32676.

NCT00125892 {published data only}

NCT00125892. A study of duloxetine in the treatment of fibromyalgia ‐ [A 1‐year safety study of duloxetine in patients with fibromyalgia; www.lillytrials.com/results/Cymbalta.pdf]. clinicaltrials.gov/show/NCT00125892; (Accessed 7 November 2013). [Lilly Clinical Trials Register Reference Summary #: 9075]

NCT00266643 {published data only}

NCT00266643. Tolerability of switching to duloxetine for the management of diabetic nerve pain (NCT00266643) [A comparison of strategies for switching patients from amitriptyline to duloxetine for the management of diabetic peripheral neuropathic pain (Lilly trial reference Summary ID# 8952).]. clinicaltrials.gov/show/NCT00266643; www.lillytrials.com/results/Cymbalta.pdf (accessed 7 November 2013). [Lilly trial reference Summary ID#: 8952]

NCT00385671 {published data only}

NCT00385671. An open‐label comparison of duloxetine to other alternatives for the management of diabetic peripheral neuropathic pain. www.clinicaltrials.gov/show/NCT00385671 (Accessed 7 November 2013).

NCT00425230 {published data only}

NCT00425230. Open label study of duloxetine for the treatment of phantom limb pain [Pilot study of use of duloxetine for the treatment of phantom limb pain]. www.clinicaltrials.org/show/NCT00425230 (accessed 7 November 2013).

NCT00552682 {unpublished data only}

NCT00552682. Pilot, opened, randomized clinical trial to assess the efficacy of duloxetine in the treatment of fibromyalgia in patients with infection by HIV 1+. www.clinicaltrials.gov\show\NCT00552682 (Accessed 7 November).

NCT00641719 {published data only}

NCT00641719. A long‐term study for the treatment of painful diabetic neuropathy. www.clinicaltrials.gov/show/NCT00641719 (Accessed 7 November 2013).

NCT01451606 {unpublished data only}

NCT01451606. Duloxetine for the treatment of chronic pelvic pain. http://clinicaltrials.gov/ct2/show/NCT01451606 (Accessed 7 November 2013). [CTG: NCT01451606]

Raskin 2005a {published data only}

Raskin J, Pritchett YL, Chappell AS, D‐Souza DN, Wong CK, Ferdinand SJ, et al. Duloxetine in the treatment of diabetic peripheral neuropathic pain ‐ results from three clinical trials. European Journal of Neurology 2005;12(s2):221.

Raskin 2006a {published data only}

Raskin J, Smith TR, Wong K, Pritchett YL, D'Souza DN, Iyengar S, et al. Duloxetine versus routine care in the long‐term management of diabetic neuropathic pain. Journal of Palliative Medicine 2006;9(1):29‐40. [PUBMED: 16430342]

Raskin 2006b {published data only}

Raskin J, Wang F, Pritchett YL, Goldstein DJ. Duloxetine for patients with diabetic peripheral neuropathic pain: a 6‐month open‐label safety study. Pain Medicine 2006;7(5):373‐85. [PUBMED: 17014595]

Russell 2006 {published data only}

Russell JW. Does duloxetine safely and effectively reduce the severity of diabetic peripheral neuropathic pain?. Nature Clinical Practice Neurology 2006;2(1):18‐9. [PUBMED: 16932514]

Skljarevski 2008 {published data only}

Skljarevski V, Desaiah D, Liu‐Siefert H, Zhang Q, Chappell AS, Detke MJ, et al. Efficacy of duloxetine in low back pain. European Journal of Neurology 2008;15(Suppl 3):320.

Skljarevski 2009 {published data only}

Skljarevski V, Ossanna M, Liu‐Seifert H, Zhang Q, Chappell A, Iyengar S, et al. A double‐blind, randomized trial of duloxetine versus placebo in the management of chronic low back pain. European Journal of Neurology 2009;16(9):1041‐8. [PUBMED: 19469829]

Skljarevski 2009a {published data only}

Skljarevski V, Desaiah D, Zhang Q, Chappell AS, Detke MJ, Gross JL, et al. Evaluating the maintenance of effect of duloxetine in patients with diabetic peripheral neuropathic pain [Maintenance of effect of duloxetine in patients with diabetic peripheral neuropathic pain]. Diabetes/Metabolism Research and Reviews 2009;25(7):623‐31. [CTG: NCT00322621; PUBMED: 19637208]

Skljarevski 2010 {published data only}

Skljarevski V, Desaiah D, Liu‐Seifert H, Zhang Q, Chappell AS, Detke MJ, et al. Efficacy and safety of duloxetine in patients with chronic low back pain. Spine 2010;35(13):E578‐85. [PUBMED: 20461028]

Skljarevski 2010b {published data only}

Skljarevski V, Zhang S, Desaiah D, Alaka KJ, Palacios S, Miazgowski T, et al. Duloxetine versus placebo in patients with chronic low back pain: a 12‐week, fixed‐dose, randomized, double‐blind trial. Journal of Pain 2010;11(12):1282‐90.

Smith 2013 {published data only}

Smith EML, Pang H, Cirrincione C, Fleishman S, Paskett ED, Ahles T, et al. Effect of duloxetine on pain, function, and quality of life among patients with chemotherapy‐induced painful peripheral neuropathy: a randomized clinical trial. JAMA 2013;309(13):1359‐67.

Tanenberg 2011 {published data only}

Tanenberg RJ, Irving GA, Risser RC, Ahl J, Robinson MJ, Skljarevski V, et al. Duloxetine, pregabalin, and duloxetine plus gabapentin for diabetic peripheral neuropathic pain management in patients with inadequate pain response to gabapentin: an open‐label, randomized, noninferiority comparison. Mayo Clinic Proceedings 2011;86(7):615‐26.

Vollmer 2011 {published data only}

Vollmer T, Robinson M, Risser R, Malcolm SK, Carlson J. A randomised, double‐blind, placebo‐controlled trial of duloxetine for the treatment of central neuropathic pain associated with multiple sclerosis. Multiple Sclerosis Journal 2011;17 (10 suppl):P1044.

Wernicke 2006b {published data only}

Wernicke JF, Raskin J, Rosen A, Pritchett YL, D'Souza DN, Iyengar S, et al. Duloxetine in the long‐term management of diabetic neuropathic pain: an open‐label, 52‐week extension of a randomized controlled clinical trial. Current Therapeutic Research 2006;67(5):283‐304.

Wu 2006 {published data only}

Wu EQ, Birnbaum HJ, Mareva MN, Le TK, Robinson RL, Rosen A, et al. Cost‐effectiveness of duloxetine versus routine treatment for U.S. patients with diabetic peripheral neuropathic pain. The Journal of Pain 2006;7(6):399‐407.

NCT00603265 {published data only}

NCT00603265. A phase 2a, randomized, double‐blind, placebo‐ and active‐controlled, parallel‐group, multicenter study to assess the safety and efficacy of ADL5859 100 mg BID in subjects with neuropathic pain associated with diabetic peripheral neuropathy [Safety and efficacy study of ADL5859 in subjects with neuropathic pain associated with diabetic peripheral neuropathy]. www.clinicaltrials.gov/show/NCT00603265 (Accessed 7 November 2013). [CTG: NCT00603265]

NCT00457730 {unpublished data only}

NCT00457730. A randomized placebo controlled trial of duloxetine for central pain in multiple sclerosis [A study to test the use of duloxetine for pain in MS]. http://clinicaltrials.gov/show/NCT00457730 Accessed 19 December 2013.

NCT00619983 {published data only}

NCT00619983. Three way interaction between gabapentin, duloxetine, and donepezil in patients with diabetic neuropathy. http://clinicaltrials.gov/show/NCT00619983 Accessed 19 December 2013.

NCT01179672 {unpublished data only}

NCT01179672. A study in patients with diabetic peripheral neuropathic pain in China. http://clinicaltrials.gov/show/NCT01179672 (Accessed 19 December 2013).

NCT01237587 {unpublished data only}

NCT01237587. Effect of duloxetine 30/60 mg once daily versus placebo in adolescents with juvenile primary fibromyalgia syndrome [A study of duloxetine in adolescents with juvenile primary fibromyalgia syndrome]. http://clinicaltrials.gov/show/NCT01237587 Accessed 19 December 2013.

NCT01552057 {unpublished data only}

NCT01552057. A phase III clinical trial of duloxetine in participants with fibromyalgia [A study of duloxetine in fibromyalgia]. http://clinicaltrials.gov/show/NCT01552057 Accessed 19 December 2013.

Aronson 2007

Aronson JK. Chapter 2 Antidepressant Drugs. In: Aronson JK editor(s). Side Effects of Drugs. Vol. 29, Amsterdam: Elsevier, 2007:23.

Aronson 2008

Aronson JK. Chapter 2 Antidepressant Drugs. In: Aronson JK editor(s). Side Effects of Drugs. Vol. 30, Amsterdam: Elsevier, 2008:19.

Attal 2006

Attal N, Cruccu G, Haanpää M, Hansson P, Jensen TS, Nurmikko T, et al. EFNS Task Force. EFNS guidelines on pharmacological treatment of neuropathic pain. European Journal of Neurology 2006;13(11):1153‐69.

Beard 2008

Beard SM, McCrink L, Le TK, Garcia‐Cebrian A, Monz B, Malik RA. Cost effectiveness of duloxetine in the treatment of diabetic peripheral neuropathic pain in the UK. Current Medical Research and Opinion 2008;24(2):385‐99.

Bellows 2012

Bellows BK, Dahal A, Jiao T, Biskupiak J. A cost‐utility analysis of pregabalin versus duloxetine for the treatment of painful diabetic neuropathy. Journal of Pain & Palliative Care Pharmacotherapy 2012;26(2):153–64.

Breivik 2006

Breivik H, Collett B, Ventafridda V, Cohen R, Gallacher D. Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. European Journal of Pain 2006;10(4):287‐333.

Burckhardt 1991

Burckhardt CS, Clark SR, Bennett RM. The fibromyalgia impact questionnaire: development and validation. Journal of Rheumatology 1991;18(5):728‐33.

Bymaster 2001

Bymaster FP, Dreshfield‐Ahmad LJ, Threlkeld PG, Shaw JL, Thompson L, Nelson DL, et al. Comparative affinity of duloxetine and venlafaxine for serotonin and norepinephrine transporters in vitro and in vivo, human serotonin receptor subtypes, and other neuronal receptors. Neuropsychopharmacology 2001;25(6):871‐80.

Bymaster 2005

Bymaster FP, Lee TC, Knadler MP, Detke MJ, Iyengar S. The dual transporter inhibitor duloxetine: a review of its preclinical pharmacology, pharmacokinetic profile, and clinical results in depression. Current Pharmaceutical Design 2005;11(12):1475‐93.

Dadabhoy 2006

Dadabhoy D, Clauw DJ. Therapy Insight: fibromyalgia ‐ a different type of pain needing a different type of treatment. Nature Clinical Practice Rheumatology 2006;2(7):364‐72.

Daousi 2004

Daousi C, MacFarlane IA, Woodward A, Nurmikko TJ, Bundred PE, Benbow SJ. Chronic painful peripheral neuropathy in an urban community: a controlled comparison of people with and without diabetes. Diabetic Medicine 2004;21(9):976‐82.

Dworkin 2008

Dworkin RH, Turk DC, Wyrwich KW, Beaton D, Cleeland CS, Farrar JT, et al. Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations. Journal of Pain 2008;9(2):105‐21.

Elbourne 2002

Elbourne DR, Altman DG, Higgins JPT, Curtin F, Worthington HV, Vail A. Meta‐analyses involving cross‐over trials: methodological issues. International Journal of Epidemiology 2002;31(1):140‐9.

Fava 2004

Fava M, Mallinckrodt CH, Detke MJ, Watkin JG, Wohlreich MM. The effect of duloxetine on painful physical symptoms in depressed patients: do improvements in these symptoms result in higher remission rates?. Journal of Clinical Psychiatry 2004;65(4):521‐30.

Gahimer 2007

Gahimer J, Wernicke J, Yalcin I, Ossanna MJ, Wulster‐Radcliffe M, Viktrup L. A retrospective polled analysis of duloxetine safety in 23,983 subjects. Current Medical Research Opinion 2007;23(1):175‐84.

GRADEpro 2008 [Computer program]

Jan Brozek, Andrew Oxman, Holger Schünemann. GRADEpro. Version Version 3.2 for Windows. Jan Brozek, Andrew Oxman, Holger Schünemann, 2008.

Hall 2006

Hall GC, Carroll D, Parry D, McQuay HJ. Epidemiology and treatment of neuropathic pain: the UK primary care perspective. Pain 2006;122(1‐2):156‐62.

Higgins 2008

Higgins JPT, Altman DG. Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S editor(s). Cochrane Handbook for Systematic Reviews of Interventions. Chichester: John Wiley & Sons Ltd, 2008:187‐244. [MEDLINE: 12251]

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Kajdasz 2007

Kajdasz DK, Iyengar S, Desaiah D, Backonja MM, Farrar JT, Fishbain DA, et al. Duloxetine for the management of diabetic peripheral neuropathic pain: evidence‐based findings from post hoc analysis of three multicenter, randomized, double‐blind, placebo‐controlled, parallel‐group studies. Clinical Therapeutics 2007;29(Supplement):2536‐46.

Mariappan 2009

Mariappan P,  Alhasso AA, Grant A, N'Dow JMO. Serotonin and noradrenaline reuptake inhibitors (SNRI) for stress urinary incontinence in adults. Cochrane Database of Systematic Reviews 2005, Issue 3. [DOI: 10.1002/14651858.CD004742.pub2]

Mease 2011

Mease PJ, Spaeth M, Clauw DJ, Arnold LM, Bradley LA, Russell IJ, et al. Estimation of minimum clinically important difference for pain in fibromyalgia. Arthritis Care & Research (Hoboken). 2011;63(8):821‐6.

Moore 2009

Moore RA, Straube S, Wiffen PJ, Derry S, McQuay HJ. Pregabalin for acute and chronic pain in adults. Cochrane Database of Systematic Reviews 2009, Issue 3. [DOI: 10.1002/14651858.CD007076.pub2]

Moore 2012

Moore RA, Derry S, Aldington D, Cole P, Wiffen PJ. Amitriptyline for neuropathic pain and fibromyalgia in adults. Cochrane Database of Systematic Reviews 2012, Issue 12. [CENTRAL: CD008242; DOI: 10.1002/14651858.CD008242.pub2]

Nagda 2004

Nagda J, Bajwa ZH. Definitions and Classification of Pain. Principles and practice of pain management. New York: McGraw Hill, 2004:51‐4.

Nose 2007

Nose M, Cipriani A, Furukawa TA, Omori I, Churchill R, McGuire H et al for the Meta‐Analysis of New Generation Antidepressants (MANGA) Study Group. Duloxetine versus other anti‐depressive agents for depression. Cochrane Database of Systematic Reviews 2007, Issue 2. [DOI: 10.1002/14651858.CD005454.pub2]

O'Connor 2008

O'Connor AB, Noyes K, Holloway RG. A cost‐utility comparison of four first‐line medications in painful diabetic neuropathy. Pharmacoeconomics 2008;26(12):1045‐64.

Perahia 2006

Perahia DG, Pritchett YL, Desaiah D, Raskin J. Efficacy of duloxetine in painful symptoms: analgesic or antidepressant effect?. International Clinical Psychopharmacology 2006;21(6):311‐7.

Saarto 2007

Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database of Systematic Reviews 2007, Issue 4. [DOI: 10.1002/14651858.CD005454]

Salaffi 2004

Salaffi F, Stancati A, Silvestri CA, Ciapetti A, Grassi W. Minimal clinically important changes in chronic musculoskeletal pain intensity measured on a numerical rating scale. European Journal of Pain 2004;8(4):283‐91.

Schuessler 2006

Scheussler B. What do we know about duloxetine's mode of action? Evidence from animals to humans. British Journal of Obstetrics and Gynaecology 2006;113 Suppl 1:5‐9.

Thorlund 2011 [Computer program]

Thorlund K, Engstrøm J, Wetterslev J, Brok J, Imberger G, Gluud C. Trial Sequential Analysis. Version Version 0.9 beta. Copenhagen: Copenhagen Trials Unit, 2011.

Treede 2008

Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, et al. Neuropathic pain: Redefinition and a grading system for clinical and research purposes. Neurology 2008;70(18):1630‐35.

Wernicke 2007

Wernicke J, Lledo A, Raskin J, Kajdasz DK, Wang F. An evaluation of the cardiovascular safety profile of duloxetine. Drug Safety 2007;30(5):437‐55.

Wiffen 2005

Wiffen PJ, McQuay HJ, Edwards JE, Moore RA. Gabapentin for acute and chronic pain. Cochrane Database of Systematic Reviews 2005, Issue 3. [DOI: 10.1002/14651858.CD005452]

Lunn 2008

Lunn MPT, Hughes RAC, Wiffen PJ. Duloxetine for treating painful neuropathy or chronic pain. Cochrane Database of Systematic Reviews 2008, Issue 2. [DOI: 10.1002/14651858.CD007115]

Lunn 2009

Lunn MPT, Hughes RAC, Wiffen PJ. Duloxetine for treating painful neuropathy or chronic pain. Cochrane Database of Systematic Reviews 2009, Issue 4. [DOI: 10.1002/14651858.CD007115.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Arnold 2004

Methods

Randomised, double‐blind, placebo‐controlled, parallel group trial of duloxetine in fibromyalgia

Participants

207 men or women over 18 years who fulfilled American College of Rheumatology criteria for fibromyalgia, and scoring 4 or more on the pain intensity item of the Fibromyalgia Impact Questionnaire (FIQ)

Interventions

Duloxetine 60 mg twice daily versus placebo for 12 weeks with a 20‐day titration phase

Outcomes

Follow‐up at 12 weeks

Outcomes:

  • FIQ pain score

  • Short Form 36 Health Survey (SF‐36)

  • Brief Pain Inventory

Notes

Greater use of antidepressants in the placebo group

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Assignment to treatment groups was determined by a computer‐generated random sequence

Allocation concealment (selection bias)

Low risk

Used an interactive voice response system

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind for all assessments in 12‐week therapy phase, Investigators adjusted the number of placebo capsules similarly to maintain the blinding. Single‐blind in run‐in phase

Incomplete outcome data (attrition bias)
All outcomes

High risk

46/104 (44%) in duloxetine and 37/103 (36%) in placebo group discontinued treatment but all dropouts accounted for and LOCF

Selective reporting (reporting bias)

Unclear risk

As above in incomplete outcome data

Other bias

Low risk

More use of antidepressants in the placebo group but this would bias against the treatment arm

Arnold 2005

Methods

Randomised, double‐blind, placebo‐controlled, parallel group trial of duloxetine in fibromyalgia

Participants

354 participants

Women only, ≥ 18 years of age who met criteria for primary fibromyalgia as defined by the American College of Rheumatology, and had a score of ≥ 4 on the average pain severity item of the Brief Pain Inventory (BPI) at randomisation

Interventions

Duloxetine 60 mg daily, duloxetine 60 mg twice daily and placebo for 12 weeks

Outcomes

  • BPI (average pain severity)

  • Short Form 36 Health Survey (SF‐36)

  • BPI interference scale

Notes

Company sponsored and run trial. Fibromyalgia Impact Questionnaire abandoned in favour of BPI

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random assignment of women who met entry criteria following the screening phase to one of three treatment groups: duloxetine 60 mg daily, duloxetine 60 mg twice daily (forced titration from 60 mg daily for 3 days to 60 mg twice daily), or placebo, with randomisation in a 1:1:1 ratio. Random assignment of the participants to treatment groups occurred within two stratified groups, those with and those without current major depressive disorder

Allocation concealment (selection bias)

Unclear risk

Probably low risk of bias as previous trial used an adequate method

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

High proportion of dropouts: 138 (39%) participants withdrew during the 12‐week therapy phase, 41 (35%) from the duloxetine 60 mg daily group, 45 (39%) from the duloxetine 60 mg twice daily group, and 52 (43%) from the placebo group (P = 0.407). Matched across groups but a high rate of loss

"Partial intention to treat analysis". Efficacy analyses include all randomised participants with a baseline and at least one post‐baseline visit with efficacy data, while safety analyses included all randomised participants

Selective reporting (reporting bias)

Unclear risk

See incomplete outcome data above

Other bias

Low risk

Lilly study. No other bias identified

Arnold 2010

Methods

Phase IV randomised, double‐blind (subject, caregiver, investigator, outcomes assessor), placebo‐controlled, parallel assignment safety and efficacy study of duloxetine in fibromyalgia

Participants

Men or women

  • Aged 18 and older who meet criteria for fibromyalgia as defined by the American College of Rheumatology

  • With a score of at least 4 on the average pain item of the Brief Pain Inventory (BPI) (modified short form) at visits 1 and 2

  • All females must test negative for pregnancy at the time of enrolment

  • A degree of understanding such that the potential participant can provide informed consent, complete protocol required assessments and communicate intelligibly with the investigator and study coordinator

Interventions

Duloxetine 60 to 120 mg daily for 24 weeks

Outcomes

Time frame for all outcome measures 24 weeks

Primary outcome

  • Patient Global Impression of Improvement (PGI‐I)

Secondary outcomes

  • BPI

  • Multidimensional Fatigue Inventory

  • Beck Depression Inventory‐II (BDI II)

  • Clinical Global Impressions of Severity (CGI‐S)

  • Beck Anxiety Inventory

  • SF‐36 (Short Form Health Survey)

  • Massachusetts General Hospital Cognitive and Physical Functioning Questionnaire

  • Anxious Likert Scale

  • Sleep Likert Scale

  • Pain Likert Scale

  • Stiffness Likert Scale

  • Mood Likert Scale

  • Columbia Suicide Severity Rating Scale

Notes

Completed and published

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned 1:1 in a double‐blind fashion to duloxetine 60 mg once daily or placebo by a computer‐generated random sequence using an IVRS

Allocation concealment (selection bias)

Low risk

"Double blind". "Variable transition to active treatment strategy…thereby blinding the onset of active treatment to reduce the patient's expectation of experiencing side effects"

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

No comment on formulation of drug or placebo but almost certainly double blinded both in up and down titration. However, significantly more participants on duloxetine withdrew with adverse effects

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Accounted for as much as possible. High dropout rate (> 30%). Employs ITT ‐ use of a "restricted maximum likelihood‐based [mixed effects model repeated measures approach] analysis accounts for bias caused by non‐random missing data due to early discontinuation because of adverse events or lack of efficacy better than LOCF"

Selective reporting (reporting bias)

Low risk

"Patient Global Impression ‐ severity (PGI‐S) only assessed at baseline". Otherwise, paper reports all results

Other bias

Unclear risk

Lilly trial. 93.2% female participants, similar to all fibromyalgia studies

Arnold 2012

Methods

Randomised, double‐blind, placebo‐controlled, parallel group study of duloxetine in fibromyalgia

Participants

Women and men > 18 years of age who met the American College of Rheumatology 1990 criteria for primary fibromyalgia and had a score of > 4 on the average pain severity item of the Brief Pain Inventory (BPI)‐Modified Short Form. Patients with or without major depressive disorder or generalised anxiety disorder, as defined by the DSM‐IV and confirmed by the MINI were included.

Interventions

Duloxetine 30 mg capsules or placebo for 12 weeks

Outcomes

Primary outcome

  • 24 hour pain severity on the BPI‐Modified Short Form

Secondary outcomes

  • Patient global impression of improvement

  • Fibromyalgia Impact Questionnaire

  • response rate (30% or 50% reduction in BPI average pain severity)

  • BPI pain severity items (pain right now, worst pain, least pain) and BPI interference score

  • Clinical Global Impression ‐ Improvement scale (CGI‐I) for depression

  • Beck Depression Inventory II

  • Beck Anxiety Inventory

  • Short Form Health Survey (SF‐36)

  • adverse events (treatment emergent, serious, vital signs and analytes, Columbia Suicide Severity Scale

Notes

Lilly study. No other bias identified

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation by a computer‐generated random sequence using an interactive voice response system (IVRS)

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Low risk

The duloxetine and placebo capsules were identical in appearance to maintain the blinding. Participants and investigators were kept blinded to the rescue criteria and dose increase; site personnel entered the major depressive disorder status at baseline and the CGI‐I for Depression scores through IVRS at every visit

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 2 dropouts, both from duloxetine group and likely to have been of minimal significance

Selective reporting (reporting bias)

Low risk

Most outcomes presented except individual BPI severity items (worst pain, least pain, pain right now). However, no other outcomes with significant effect in completely negative trial

Other bias

Low risk

Lilly study. No other bias identified

Brecht 2007

Methods

8‐week, randomised, double‐blind, placebo‐controlled, parallel‐group efficacy and safety study of duloxetine in the treatment of pain of unknown aetiology in people with major depressive disorder

Participants

Women or men > 18 with major depressive disorder defined by DSM‐IV. At baseline, depression score of > 20 on the MADRS and at least moderate pain on Brief Pain Inventory Short Form (BPI‐SF) ‐ 3 or higher for "24 hour average pain". Participants were also devoid of any other diagnosed pain syndrome as per a medical history

Interventions

Duloxetine 60 mg versus placebo for 8 weeks

Outcomes

Primary outcome

  • Mean change in the BPI‐SF 24 hour pain during 8 weeks of treatment

Seconday outcomes

  • Response rates to individual BPI severity items (worst pain, least pain, pain right now), and interference items (30% or more from baseline and sustained if maintained response from response definition to 8‐week completion)

  • MADRS (max score 60, reduction of 50% defined as response, sustained as above)

  • Clinical Global Impression ‐ Severity scale (CGI‐S)

  • Clinical Global Impression ‐ Improvement scale (CGI‐I)

  • Patient Global Impression of Improvement (PGI‐I)

  • The Symptom Checklist‐90‐R (SCL‐90‐R),

  • adverse events, treatment emergent adverse events, vital signs, laboratory parameters

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomised"

Allocation concealment (selection bias)

Unclear risk

"Double blind"

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

"Double blind"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT using all participants with 1 dose of drug. 25% dropout rate

Selective reporting (reporting bias)

Low risk

None identified

Other bias

Low risk

Company sponsored trial

Chappell 2008

Methods

Six‐month, randomised, double‐blind, placebo‐controlled, clinical trial of duloxetine in fibromyalgia

Participants

Male and female outpatients were eligible for the study if they were ≥ 18 years of age, met criteria for fibromyalgia as defined by the American College of Rheumatology, with or without major depressive disorder

No criteria for pain level at entry

Interventions

Duloxetine ‐ variable dose. Started at 60 mg (30 mg run in period over 1 week), randomised increase to 120 mg after 13 weeks if not > 50% reduction in pain on BPI average

Outcomes

  • BPI‐I at > 12 weeks. No data given for less than 12 weeks although "statistically significant" P values quoted without figures at weeks 1, 2, 4, 6 and 8 BUT NOT 13, then week 18

  • Short Form Health Survey (SF‐36)

  • Patient Global Impression of Improvement

  • Fibromyalgia Impact Questionnaire (FIQ)

  • Clinical Global Impression ‐ Severity scale (CGI‐S)

  • Multidimensional fatigue inventory

  • Hospital Anxiety and Depression Scale (HADS)

  • Hamilton Depression Rating Scale (HAMD)

  • Beck Depression Inventory –II

  • Sheehan Disability Scale

  • EQ‐5D

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer generated random sequence within each study centre stratified by major depressive disorder

Allocation concealment (selection bias)

Low risk

Double‐blind

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Participants "blinded", but not clear how the study managed dose escalations and decreases and whether blinding was maintained

Incomplete outcome data (attrition bias)
All outcomes

High risk

37.6% to 38.6% discontinuations, significantly different in lack of efficacy only. Investigators used LOCF and MMRM to correct for dropouts

Selective reporting (reporting bias)

Low risk

30% improvement in BPI‐average added post hoc

Other bias

Unclear risk

Lilly sponsored trial

Significant unexplained treatment by investigator interaction

Gao 2010

Methods

Phase III randomised, double‐blind (subject, caregiver, investigator, outcomes assessor), placebo‐controlled, parallel assignment safety and efficacy study of duloxetine in painful diabetic neuropathy

Participants

215 participants

Men or women, aged 18 to 75, pain due to bilateral peripheral neuropathy caused by type I or type II diabetes with the pain beginning in the feet, and present for at least 6 months. Score of 4 or greater on the Brief Pain Inventory (BPI) on the 24‐hour average pain item

Interventions

Duloxetine 60 mg daily for 12 weeks

Outcomes

Primary outcome

  • BPI 24‐hour average pain score (efficacy of duloxetine 60 to 120 mg daily)

Secondary outcomes

  • BPI worst pain, least pain, and current pain severity and average of 7 interference scores

  • Clinical Global Impression of Severity

  • Patient Global Impression of Improvement

  • EuroQoL Questionnaire ‐ 5 dimensions

  • Discontinuation rates

  • Tolerability of morning versus evening dosing, spontaneously reports adverse events

  • Athens Insomnia Scale 8‐item and 5‐item

  • Adverse events

  • Vital signs

  • Laboratory measures

Notes

Closed and completed

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned

Allocation concealment (selection bias)

Low risk

"Double blind". Study medication in capsules…or matching placebo'

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

15.6% to 17.9% dropout rate but ITT using LOCF and MMRM approach to minimise bias

Selective reporting (reporting bias)

Low risk

Selective use of MMRM or LOCF depending upon outcome. However, all measures reported

Other bias

Low risk

Lilly sponsored trial

No adjustment for multiple comparisons

Gaynor 2011a

Methods

Randomised, double‐blind, placebo‐controlled trial of duloxetine in people with major depressive disorder and painful physical symptoms

Participants

Adult (18 years of age) male or female outpatients were eligible ...if they met all of the following: a current episode of major depressive disorder according to the DSM‐IV‐TR and confirmed by the MINI with a history of at least one separate, previous episode of depression, and at both the screening and randomisation visits a MADRS total score of 20, and at least moderate pain with a score of 3 on the Brief Pain Inventory Short Form (BPI) average pain item, and a Clinical Global Impression of Severity (CGI‐S) score 4. Painful symptoms were not allowed to have an identifiable underlying cause

Interventions

Duloxetine 60 mg once daily orally for 8 weeks vs placebo

Outcomes

  • BPI at 8 weeks

  • Patient reported global impression of improvement

  • Sheehan Disability Scale global functional impairment score

Notes

Gaynor 2011b identical in design ‐ different patient group

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No concerns

Selective reporting (reporting bias)

Low risk

None

Other bias

Low risk

None

Gaynor 2011b

Methods

A randomised, double‐blind, placebo‐controlled trial of duloxetine in people with major depressive disorder and painful physical symptoms

Participants

Adult (18 years of age) male or female outpatients were eligible ...if they met all of the following: a current episode of major depressive disorder according to the DSM‐IV‐TR and confirmed by the MINI with a history of at least one separate, previous episode of depression, and at both the screening and randomisation visits a MADRS total score of 20, and at least moderate pain with a score of 3 on the Brief Pain Inventory Short Form (BPI) average pain item, and a Clinical Global Impression of Severity (CGI‐S) score 4. Painful symptoms were not allowed to have an identifiable underlying cause

Interventions

Duloxetine 60 mg once daily orally for 8 weeks vs placebo

Outcomes

  • BPI at 8 weeks

  • Patient reported global impression of improvement

  • Sheehan Disability Scale global functional impairment score

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

'Randomised'

Allocation concealment (selection bias)

Unclear risk

Unclear

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

'Double Blind'

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No concerns

Selective reporting (reporting bias)

Low risk

None

Other bias

Low risk

None identified

Goldstein 2005

Methods

Randomised, double‐blind, placebo‐controlled, parallel group, trial of duloxetine in painful diabetic neuropathy

Participants

457 participants

Participants, at least 18 years of age, had daily pain due to polyneuropathy caused by type 1 or type 2 diabetes mellitus, which was present for a minimum of 6 months. This pain had to have begun in the feet with relatively symmetrical onset. The diagnosis was confirmed by a score of at least 3 on the Michigan Neuropathy Screening Instrument (MNSI). Participants were required to have a minimum score of 4 on the 24‐hour average pain score rated on an 11‐point (0 to 10) Likert scale.

Interventions

Duloxetine 20 mg daily, 60 mg daily or 60 mg twice daily versus placebo for 8 weeks

Outcomes

  • 24‐hour average pain score

  • SF‐36

  • Patient Global Impression of Change,

  • Night pain

Notes

Company sponsored and run trial

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned in a 1:1:1:1 ratio by a computer generated random sequence

Allocation concealment (selection bias)

Low risk

Participant numbers were assigned consecutively at each study site. The interactive voice response system was used to assign blister cards containing the study drug to each participant confirmed through interactive voice response system entry of a confirmation number on the card

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

All analyses were undertaken as an ITT analysis. All participants were analysed in the safety analysis and all participants with at least one post entry data point were analysed in an ITT analysis. Dropout rate was 25% with significantly more in the higher dose treatment groups

Selective reporting (reporting bias)

Unclear risk

See above

Other bias

Low risk

Company sponsored and run trial

Kaur 2011

Methods

Randomised, double‐blind, cross‐over clinical trial comparing amitriptyline and duloxetine in painful diabetic neuropathy

Participants

86 participants ‐ 65 randomised to treatment in 1st arm, 58 of whom completed both arms

People of either sex with type 2 diabetes, aged between 18 and 75 years, who were on stable glucose‐lowering medications during the preceding month and who had painful diabetic neuropathy for at least 1 month were considered for the study. The study enrolled people who had a pain score of > 50%, as assessed by visual analogue scale (VAS). Painful diabetic neuropathy was confirmed by 1) medical history, 2) a diabetic neuropathy symptom (DNS) score of > 1 point (7), 3) a Diabetic Neuropathy Examination (DNE) score of > 3 points (8), 4) a modified neuropathy symptom score (mNSS) (9,10), and 5) increased thresholds on the vibration perception test and monofilament test

Interventions

Amitiyptyline 10, 25 or 50 mg once daily at night or duloxetine, 20, 40 or 60 mg once daily at night

Intervention only 6 weeks before 2 week washout and cross‐over to alternate arm. Participants commenced on lowest dose and then increased every 2 weeks to next dose if required by treating physician; thus potentially only 2 weeks on maximum dose. 48% of amitriptyline and 65% of duloxetine participants reached the highest dose of drug. 17% vs 5% of the participants preferred higher dose duloxetine to amitriptyline

Outcomes

Primary outcome

  • Patient's global assessment of efficacy by VAS (0 to 100)

Secondary outcomes

  • Short form McGill pain questionnaire (11)

  • 11‐point Likert scale for pain (0 to 10)

  • DNE score

  • DNS score

  • mNSS

  • Hamilton Depression Rating Scale (HAMD)

  • change in sleep pattern (increased, unchanged or decreased)

  • Patient global impression of change

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised (computer generated randomisation of blocks of 4)

Allocation concealment (selection bias)

Unclear risk

An independent person unrelated to the study carried out blinding and randomisation. Two separate companies provided medicines, so it is not clear that they were identical in appearance

Blinding (performance bias and detection bias)
All outcomes

Low risk

Blinded. Single physician assessment. "success of blinding was assessed by the accuracy of the physicians prediction at the end of the study" (34% correctly identified only)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No concerns

Selective reporting (reporting bias)

High risk

The primary end point of the study was the reduction in the median pain score from baseline, (patient’s global assessment of efficacy by VAS (0 to 100 points)). Secondary end points included the assessment of pain by the short‐form McGill Pain Questionnaire (11); an 11‐point Likert scale for pain (0 = no pain and 10 = excruciating pain); change in sleep pattern (increased, unchanged, or decreased); overall improvement by DNE score, DNS score, mNSS, and the 24‐point HAMD; and patient self evaluation of overall change on the basis of a 7‐point Patient Global Impression of Change (PGIC) scale not reported in analysis.

Other bias

High risk

Significant (but similar) carryover into period 2 despite 2 weeks' washout

Raskin 2005

Methods

Randomised, double‐blind, placebo‐controlled, parallel group trial in diabetic peripheral neuropathic pain

Participants

348 participants

Participants ≥ 18 years, with pain due to bilateral peripheral neuropathy caused by type 1 or type 2 diabetes mellitus. The pain had to begin in the feet with relatively symmetrical onset and be present for at least 6 months. Participants had to have a mean score of ≥ 4 when assessed for 24‐hour average pain severity on the Michigan Neuropathy Screening Instrument (MNSI) 11‐point Likert scale (from the patient diary prior to randomisation), and stable glycaemic control. Concomitant pain medications excluded.

Interventions

Duloxetine 60 mg daily or duloxetine 60 mg twice daily versus placebo for 12 weeks

Outcomes

  • 24‐hour average pain severity

  • Patient global impression of clinical change,

  • pain at rest

  • Brief Pain Inventory (BPI) severity

  • Clinical Global Impression of Pain Severity scale (CGI‐S)

  • Short Form McGill pain questionnaire

  • BPI interference scale

Notes

Company sponsored and run trial

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation performed at visit 3 in a 1:1:1 ratio. A computer‐generated random sequence determined assignment to treatment groups, using an IVRS

Allocation concealment (selection bias)

Low risk

Participants received either of (or a combination of, depending on their randomly assigned treatment) the following: 30 mg capsules of duloxetine hydrochloride or placebo capsules identical to duloxetine capsules. Participants randomly assigned to each treatment group were instructed to take two capsules (by mouth) every morning and every evening.

Treatment was assigned using IVRS

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts were 52/340 (15%). Analysis was by ITT

Selective reporting (reporting bias)

Low risk

See above

Other bias

Low risk

Lilly study. No other bias identified

Rowbotham 2012

Methods

Phase II, randomised, double‐blind, placebo‐controlled, single group assignment, safety and efficacy study comparing duloxetine, ABT‐894 and placebo in diabetic neuropathic pain

Participants

108 participants

Men and women 18 to 75

Inclusion criteria:

  • The subject must have a diagnosis of diabetes mellitus (type 1 or type 2) and a diagnosis of diabetic neuropathic pain

  • Participant's diabetic neuropathic pain must be present for a minimum of six months and should have begun in the feet with relatively symmetrical onset.

  • Participant has an HbA1c ≤ 9. Participants who have an HbA1c > 9 and ≤11 may be included in the study

Interventions

Drug: ABT‐894, 1 mg, 2 mg, 4 mg twice daily
Drug: placebo
Drug: duloxetine 60 mg

Duration 8 weeks

Outcomes

Primary outcome

  • Efficacy of each ABT‐894 dose (1 mg, 2 mg or 4 mg twice daily) versus placebo in the treatment of pain due to diabetic neuropathic pain (time frame: change from baseline to final 24‐hour average pain score)

Secondary outcomes

  • Proportions of treatment responders; subjects who complete treatment period with 30% improvement (time frame: from baseline to final 24‐hour average pain score)

  • Mean of 24‐hour worst pain severity, average of night pain, and average of morning pain measured by the 11‐point Likert scale and from participant's daily diary (time frame: weekly during treatment)

  • BP‐(Short Form) including pain severity (time frame: at each visit from baseline to week 8)

  • Clinician Global Impression Severity scale (CGI‐S) and Patient Global Impression of Change (PGIC) (time frame: at each visit from baseline to week 8) 

Notes

Published 2012

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants "were randomized 1:1 to each treatment arm via an interactive voice response system using a randomization schedule that was generated before study start"

Allocation concealment (selection bias)

Low risk

Careful attention to placebo and medication concealment noted

Blinding (performance bias and detection bias)
All outcomes

Low risk

No concerns

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Almost 100% completed

Selective reporting (reporting bias)

Low risk

All reported

Other bias

Low risk

None identified

Russell 2008

Methods

Randomised, double‐blind, placebo‐controlled, parallel group trial in fibromyalgia

Participants

520 participants

Female and male outpatients ≥ 18 years of age who met criteria for fibromyalgia as defined by the American College of Rheumatology. Participants were required to have a score ≥ 4 on the average pain severity item (in the past 24 hours) of the Brief Pain Inventory (BPI‐modified Short Form at screening and at baseline. The study included people with or without current major depressive disorder and evaluated them for the presence of psychiatric disorders using the MINI. Prior to randomisation, the study required participants to discontinue any medications that might interfere with the evaluation of pain improvement, including analgesics (with the exception of up to 325 mg/day of aspirin for cardiac prophylaxis and paracetamol up to 2 g/day for pain), antidepressants, anticonvulsants, or other medications taken for fibromyalgia or pain

Interventions

Duloxetine 20 mg daily, 60 mg daily or 60 mg twice daily versus placebo for 6 months

Outcomes

  • BPI average pain severity score

  • Short Form 36 Health Survey (SF‐36)

  • patient global impression of clinical change

Notes

Company sponsored and run trial

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A computer‐generated random sequence determined assignment to treatment groups and the study randomly assigned each stratum (depressed and non‐depressed) within sites to achieve a relative balance across treatments

Allocation concealment (selection bias)

Unclear risk

Unclear although other trials from the same group have been adequate

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

35% to 40% dropout at the 3 month interim analysis phase and up to 46% dropout for the 6 month phase. "Intention‐to‐treat unless otherwise specified". Safety analyses in all participants and others with data for at least 1 measure

Selective reporting (reporting bias)

Unclear risk

See above

Other bias

Low risk

Lilly study. No other bias identified

Tesfaye 2013

Methods

Randomised, double‐blind, placebo‐controlled, parallel group enrichment trials with three phases comparing duloxetine to pregabalin in painful diabetic neuropathy

Participants

401 participants treated with duloxetine and 403 with pregabalin

Included participants had pain due to bilateral peripheral neuropathy (caused by type 1 or type 2 diabetes mellitus. Pain must have begun in the feet, with relatively symmetrical onset. Daily pain should have been present for more than 3 months (assessed by questioning the patient).
• Score of at least 4 on the 24‐hour average pain severity score on an 11‐point Likert scale (on Brief Pain Inventory Modified Short Form (BPI‐mSF)) at screening and at randomisation
• Participants not receiving treatment for diabetic peripheral neuropathic pain or received treatment for diabetic peripheral neuropathic pain, with a drug other than pregabalin or duloxetine, and completed the required washout
• Participants never received treatment with duloxetine or pregabalin (short courses of less than 15 days of treatment, at any time previously, allowed)
•Stable glycaemic control, as assessed by a physician investigator, and HbA1c less than or equal to 12% at screening

Interventions

Pregabalin titrated to 150 mg twice daily was compared to duloxetine titrated to 60 mg once daily (with placebo tablets to maintain blind between treatments) and treated in study phase II for 8 weeks. A third phase of non‐responding participants entered study phase III not included in this analysis

Outcomes

Primary outcome

  • 24‐hour average pain on BPI‐mSF VAS. Response rates of 30%, 50% or 2‐point reduction collected at all visits

Secondary outcomes

  • BPI‐SF items as other studies

  • Clinical Global Impression ‐ Improvement scale (CGI‐I)

  • Patient Global Impression ‐ Improvement (PGI‐I)

  • Neuropathic Pain Symptom Inventory (NPSI) and 5 subscores

  • HADS

  • 24‐hour average pain on the BPI‐mSF for period 2 of trial (initial therapy)

  • Treatment emergent adverse events, serious adverse events, vital signs, laboratory values, Beck Depression Inventory II (BDI‐II) to assess suicide risk

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised 1:1:1:1 in 4 parallel groups based on a computer generated sequence using IVRS

Allocation concealment (selection bias)

Unclear risk

Unclear ‐ although all drugs and placebo were similar and the allocation stratified by site, does not explicitly deal with concealment

Blinding (performance bias and detection bias)
All outcomes

Low risk

The trial maintained blinding by using over‐encapsulated duloxetine and pregabalin capsules, matching placebo and an identical dosing regimen for all groups in terms of numbers and timing of capsules

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Dropout in Phase II 17%, 9% with adverse events. All analyses performed on ITT (baseline + 1 measure for outcomes, all randomised for adverse events) with MMRM ‐ however no statement as to whether LOCF or BOCF used

Selective reporting (reporting bias)

High risk

Some partial reporting of outcomes (for example NPSI subscores not tabulated, PGI‐I and CGI‐I in figure form only and differences of reporting between phase II and phase III outcome reporting

Other bias

High risk

Lilly designed, interpreted, wrote and submitted. Ghost written by professional writer for company

Vranken 2011

Methods

Stratified, randomised, double‐blind, placebo‐controlled, parallel group study of patients with severe central neuropathic pain of more than 6 months duration from cerebrovascular or spinal cord lesions

Participants

48 participants aged 18 years or older with > 6 month severe neuropathic pain from cord or cerebrovascular cause, > 6 on visual analogue scale (VAS) (10 points), which started after sustaining the lesion and with the distribution of pain concomitant with the somatosensory system involvement. The trial allowed other medication if doses were stable for 6 weeks, except other antidepressants, which had to be stopped more than 30 days prior to receiving study medication

Interventions

Duloxetine or placebo for 8 weeks. Duloxetine 60 mg at start. Increased if participants did not meet criteria of > 1.8 points improvement on VAS. At week 8 and study end 15 participants on 120 mg and 8 participants on 60 mg

Outcomes

Primary outcome

  • Pain intensity on a 10‐point VAS measured a baseline and weekly for the 8 weeks of the study. The final mean pain score was an average of 9 VAS scores measured over 72 hours in the last 3 days of the study

Secondary outcomes

  • Pain disability Index

  • EQ‐5D

  • Short Form 36 Health Survey (SF‐36) (beginning and end)

  • Patient Global Impression of Change (PGIC) (end of study only)

  • Quantitative sensory testing

  • Adverse events

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Simple computerised random sampling (clorandm.exe) assigned study codes N = 1 to the placebo or duloxetine arm. Consecutive participants who met inclusion criteria were randomly assigned to treatment with flexible dose placebo or flexible dose duloxetine

Allocation concealment (selection bias)

Low risk

The association between type of treatment and study code was only known to the Department of Epidemiology, Biostatistics and Bioinformatics and the hospital pharmacy department

Blinding (performance bias and detection bias)
All outcomes

Low risk

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Selective reporting (reporting bias)

Low risk

Other bias

Low risk

Wernicke 2006

Methods

Randomised, double‐blind, placebo‐controlled, parallel group trial of duloxetine in diabetic peripheral neuropathic pain

Participants

334 participants

Men or women ≥ 18 years and with > 6 months diabetic peripheral neuropathic pain secondary to type 1 or 2 diabetes (distal and symmetrical). At randomisation, score > 3 on Michigan Neuropathy Screening Instrument and average > 4 on 24 hour pain scale. Stable glucose control and HBA1c < 12. Multiple exclusions including other pain medications except paracetamol and aspirin

Interventions

Duloxetine 60 mg daily, 60 mg twice daily or placebo for 12 weeks

Outcomes

Primary outcome

  • 24 hour average pain score (Likert 11‐point)

Secondary outcomes

  • SF‐36

  • BPI interference,

  • patient reported global clinical impression of change,

  • night pain,

  • clinical global impression ‐ pain severity (CGI‐S),

  • clinical global impression of change

Notes

Company sponsored and run trial

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was performed at the site level in that randomisation codes were assigned to sites in blocks, but there was no further stratification. Participants were randomly assigned to treatment in a 1:1:1 ratio. Assignment to a treatment group was determined by a computer‐generated random sequence using an IVRS

Allocation concealment (selection bias)

Low risk

The IVRS was used to assign blister cards containing study drug to each participant

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

Drop outs were 29/114 (25%) in duloxetine 60 mg daily, 34/112 (30%) in duloxetine 60 mg twice daily and 23/108 (21.3%) in the placebo group

Selective reporting (reporting bias)

Unclear risk

An ITT principle was used in the analyses of all efficacy variables. For each efficacy variable, the analysis included all randomised participants with a baseline and at least one non‐missing observation after baseline

Other bias

Low risk

Lilly study. No other bias identified

Yasuda 2010

Methods

Phase III randomised, double‐blind (subject, caregiver, investigator, outcomes assessor), placebo‐controlled, parallel assignment, safety and efficacy study of duloxetine in diabetic peripheral neuropathic pain

Participants

339 participants randomised

Male or female outpatients aged 20 years or older but less than 80 years at the time of consent:

  • with pain due to bilateral peripheral neuropathy induced by type 1 or 2 diabetes mellitus. The pain must have been present for at least 6 months and be evaluable in feet, legs, or hands

  • with HbA1c less than or equal to 9.0 percent at visit 1

  • in whom HbA1c had been measured 42 to 70 days before visit 1 and subsequent HbA1c levels have been within +/‐ 1.0 percent of the level at visit 1

  • with a mean of the 24‐hour average pain severity scores (round off to a whole number) of 4 or higher, as calculated from the patient diary for 7 days immediately before visit 2

Interventions

Duloxetine 40 mg or 60 mg orally daily versus placebo for 12 weeks

Outcomes

Primary outcome

  • Reduction in average pain severity as measured by an 11‐point Likert scale (time frame 12 weeks)

Secondary outcomes

  • Pain severity for worst pain and night pain as measured by an 11‐point Likert scale (time frame 3 months)

  • Patient Global Impression of Improvement scale to measure the degree of improvement at the time of assessment (time frame 3 months) 

  • Brief Pain Inventory to measure the severity of pain (time frame 3 months)

  • Beck Depression Inventory‐II (BDI‐II) total score (time frame 3 months)

  • Safety (time frame 3.5 months)

Notes

Recruiting

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Assigning table was prepared using Create Key Code 3.3. Participants were randomly assigned…' Stratified for pain, duration of diabetic peripheral neuropathy, diabetes type, study centre

Allocation concealment (selection bias)

Unclear risk

No clear explanation of methodology

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

"Double blind"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

10.2% to 16.9% dropout. All analyses using LOCF and MMRM

Selective reporting (reporting bias)

Low risk

None identified

Other bias

Low risk

Lilly sponsored

DSM‐IV: Diagnostic and Statistical Manual of Mental Disorders, 4th ed
HbA1c: haemoglobin A1c
ITT: intention‐to‐treat
BOCF: best observation carried forward
LOCF: last observation carried forward
MADRS: Montgomery–Åsberg Depression Rating Scale
MINI: Mini International Neuropsychiatric Interview
MMRM: mixed‐effect model repeated measure

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Boyle 2012

28 days only

Brannan 2005

6 weeks of treatment only

Canovas 2007

Not randomised or controlled

Chappell 2009

Osteoarthritis of the knee ‐ likely to cross over with Cochrane Musculoskeletal Group

Chappell 2011

Osteoarthritis of the knee ‐ likely to cross over with Cochrane Musculoskeletal Group

Goldstein 2004

Trial of duloxetine in depression. Pain scales as secondary outcome measures only. It was not clear what sort of pain the participants had (for example musculoskeletal, neuropathic, headache) and the levels of pain at baseline were low compared to the included trials

Harrison 2013

Four weeks treatment only in each group of 4 way crossover. Terminated early July 2013

Lavoie Smith 2012

Abstract publication of Smith 2013

NCT00125892

Open and then double‐blind study comparing 2 doses of duloxetine 60 mg and 120 mg

NCT00266643

The first part of this cross‐over study was the only part of trial suitable for assessment (amitriptyline versus duloxetine) but only 4 weeks long ‐ thus excluded

NCT00385671

Open label

NCT00425230

Was registered in clinicaltrials.gov ‐ study terminated with no participants enrolled because no drug supplied

NCT00552682

Open label duloxetine vs no treatment or pre‐existing antidepressant

NCT00641719

Open label extension of Yasuda 2010

NCT01451606

Pelvic pain

Raskin 2005a

Not a randomised controlled study but a report of 3 trials included in this review

Raskin 2006a

Not a double‐blind trial

Raskin 2006b

Open label study with dosage control only

Russell 2006

Summary report of 3 studies included in this review

Skljarevski 2008

Back pain ‐ to be included in a Cochrane Back Group review ‐ Back Group informed. Published in full format in 2009 European Journal of Neurology 16: 1041‐8

Skljarevski 2009

Back pain ‐ to be included in a Cochrane Back Group review ‐ Back Group informed

Skljarevski 2009a

Open label extension

Skljarevski 2010

Back pain ‐ to be included in a Cochrane Back Group review ‐ Back Group informed

Skljarevski 2010b

Back pain ‐ to be included in a Cochrane Back Group review ‐ Back Group informed

Smith 2013

Duration of treatment only 4 weeks

Tanenberg 2011

Open label ‐ non blinded study

Vollmer 2011

Measured outcomes at durations of less than eight weeks

Wernicke 2006b

Not double‐blind ‐ extension of Goldstein 2005

Wu 2006

Open study, not blinded

Characteristics of studies awaiting assessment [ordered by study ID]

NCT00603265

Methods

Phase II randomised, double blind, parallel assignment, safety and efficacy study

Participants

Male and female participants between 18 and 75 years of age

Diabetes mellitus (type I or II) that is documented to be under stable glycaemic control over a period of at least 3 months, as indicated by a HbAIc of ≤ 12% and a stable dose of insulin or oral diabetic medication for 90 days prior to starting study medication. Evidence of symmetrical, bilateral pain in the lower extremities due to diabetic peripheral neuropathy. Presence of daily pain due to DPN for at least 3 months. Score ≥ 3 on the physical examination portion of the Michigan Neuropathy Screening Instrument (MNSI). Average weekly pain score of ≥ 4 on the numeric pain rating scale (NPRS) for symmetrical neuropathic pain in the feet and legs

Interventions

Drug: ADL5859
Drug: duloxetine
Drug: placebo

Outcomes

Primary outcome measures:

  • Change from baseline in mean NPRS (time frame: week 4)

Secondary outcome measures:

  • Change from baseline in the mean NPRS proportion of subjects with 30% reduction in average pain score (weekly)

  • Patient Global Impression of Change (PGIC) (time frame: week 4)

  • Change in Sleep Interference Scale (SIS) from baseline (time frame: week 4)

  • Change from baseline in the evening assessment of the 24‐hour overall mean pain intensity score (time frame: weekly)

  • Change from baseline in NPRS at rest in the clinic (time frame: weekly)

  • Change from baseline in NPRS after walking 50 feet in the clinic (time frame: weekly)

Notes

Completed ‐ no reference in Pubmed ‐ no information on clinicaltrials.gov‐ e‐mail written to company with request for information September 2012.

HbA1c: haemoglobin A1c

Characteristics of ongoing studies [ordered by study ID]

NCT00457730

Trial name or title

A randomised placebo controlled trial of duloxetine for central pain in multiple sclerosis

Methods

Randomised, double‐blind (caregiver, investigator), placebo‐controlled, parallel assignment safety/efficacy study

Participants

People with multiple sclerosis "who have central pain which is 4 or greater on a scale of 1‐10. Patients must have experienced pain for 2 months or longer prior to beginning the study."

Interventions

Duloxetine 30 mg (10 capsules) for 1 week, titrated up to 60 mg (40 capsules) for 5 weeks and titrated back down to 30 mg for 1 week

Placebo for 7 weeks

Outcomes

Time frame for all outcomes, week 2 and week 6

  • Weekly means of:

    • 24 hour average pain score

    • 24 hour worst pain score

    • sleep rating

  • Global Impression of Change

  • SF‐36

  • Beck Depression Inventory

  • Average daily consumption of ibuprofen

Starting date

January 2007

Contact information

Brown, Theodore R., M.D., MPH

Evergreen Healthcare Kirkland, Washington, United States, 98034

Notes

NCT00457730 Lilly sponsored

NCT00619983

Trial name or title

Three way interaction between gabapentin, duloxetine, and donepezil in patients with diabetic neuropathy

Methods

Randomised, double‐blind (subject, investigator, outcomes assessor), parallel assignment

Participants

Male or female. Diagnosis of diabetic neuropathy. Age 18 to 80

Interventions

Group 1: donepezil 5 mg once per day for 12 weeks

Group 2: duloxetine 30 mg twice a day for 12 weeks

Group 3: combination of donepezil 2.5 mg and duloxetine 30 mg for 12 weeks

Group 4: placebo pills.

Gabapentin added to all groups at week 9

Outcomes

Primary:

  • Pain intensity measurements recorded twice daily, using McGill short form pain questionnaire on a handheld computer. The Visual Analog Pain Scale (VAS) served as the primary outcome measure (time frame: study completion (16 weeks)

Starting date

February 2008 to July 2010

Contact information

Regina Curry, RN, CCRC

336‐716‐4294

[email protected]

Wake Forest University Baptist Medical Center

Winston‐Salem, North Carolina, United States, 27157

Notes

NCT00619983 Still recruiting 2013 ‐ estimated completion July 2013

NCT01179672

Trial name or title

Treatment of patients with diabetic peripheral neuropathic pain in China: duloxetine versus placebo

Methods

Randomized, double blind (subject, investigator), placebo‐controlled, parallel assignment, efficacy study

Participants

People over 18 years of age who present with pain due to bilateral diabetic peripheral neuropathy (type 1 or type 2 diabetes). Pain beginning in feet, relatively symmetrical onset, present daily for at least 6 months, confirmed by score of ≥ 3 on Michigan Neuropathy Screening Inventory

Interventions

Duloxetine 30 mg orally, once daily for 1 week; 60 mg once daily for next 11 weeks; 30 mg administered orally, once daily for 1 week during taper period

Placebo once daily for 12 weeks, once daily for 1 week during taper period

Outcomes

Primary:

  • Mean change in the pain severity score (measured from baseline to 12‐week endpoint)

Secondary (changes measured from baseline to 12‐week endpoint):

  • Mean change in night pain and worst pain

  • Mean change in the Brief Pain Inventory (BPI)‐Severity scale

  • Mean change in the Clinical Global Impression ‐ Severity (CGI‐S) scale

  • Patient Global Impression of Improvement (PGI‐I) scale

  • Mean change in the Sensory portion of the Short‐form McGill pain questionnaire

  • Percentage of participants who experience ≥ 30%, ≥ 50% or ≥ 75% reduction from baseline to 12 week endpoint in average daily pain

  • Percentage of participants who experience ≥ 30%, ≥ 50% or ≥ 75% reduction from baseline in BPI‐Severity average pain scores

  • Mean change in the Brief Pain Inventory (BPI) Interference scores

  • Mean change in the Sheehan Disability Scale (SDS)

Starting date

April 2011

Contact information

Eli Lilly and Company. Study director, tel: 1‐877‐CTLILLY (1‐877‐285‐4559) or 1‐317‐615‐4559

Notes

NCT01179672

NCT01237587

Trial name or title

A study of duloxetine in adolescents with juvenile primary fibromyalgia syndrome

Methods

Phase III, randomised, double‐blind (subject, caregiver, investigator, outcomes assessor), parallel assignment, safety/efficacy study

Participants

Aged 13 to 17 years who meet criteria for primary juvenile primary fibromyalgia syndrome and have a score of greater than or equal to 4 on Brief Pain Inventory (BPI) average pain severity (Item 3) during screening

Interventions

Blinded period: 30 mg or 60 mg duloxetine or placebo once daily for 13 weeks

Open label extension: 30 mg or 60 mg duloxetine once daily for 26 weeks

Outcomes

Primary:

  • Change from baseline to 13 week endpoint in Brief Pain Inventory (BPI) modified short form‐adolescent version 24 hour average pain severity item

Secondary:

  • Change from baseline to endpoint (13 weeks, 39 weeks extension phase) in Brief Pain Inventory (BPI) modified short form‐adolescent version severity and interference items

  • Maintenance effect in acute phase responders on the Brief Pain Inventory (BPI) modified short form‐adolescent version 24 hour average pain severity item (endpoint 13 weeks, 39 weeks extension phase)

  • Proportion of participants with ≥ 30% and ≥ 50% reduction in BPI 24 hour average pain severity score at 13 weeks

  • Change from baseline (endpoint 13 weeks, 39 weeks extension phase) in:

    • Pediatric Pain Questionnaire (PPQ) item scores

    • Clinical Global Impression ‐ Severity (CGI‐S): overall score and mental illness score

    • Functional Disability Inventory (FDI) child scale and rent scale

    • Children's Depression Inventory (CDI)

    • Multidimensional Anxiety Scale for Children (MASC)

Starting date

February 2011

Contact information

Eli Lilly and Company. Study Director: 1‐877‐CTLILLY (1‐877‐285‐4559) or 1‐317‐615‐4559

Notes

NCT01237587

NCT01552057

Trial name or title

A phase III clinical trial of duloxetine in participants with fibromyalgia

Methods

Randomised, double‐blind (subject, investigator), placebo‐controlled, parallel assignment, safety/efficacy study

Participants

Participants with fibromyalgia aged 20 to 74 years

Inclusion criteria:

  • fulfilling American College of Rheumatology 1990 criteria for fibromyalgia

  • pain severity ≥ 4 by Brief Pain Inventory (BPI) ‐ average pain severity item (question 3)

Interventions

Duloxetine hydrochloride orally 60 mg for 15 weeks or oral placebo for 15 weeks

Outcomes

Changes measured from baseline to 14 week endpoint

Primary:

  • 24‐Hour Average Pain Severity Item of the BPI‐Modified Short Form Score

Secondary:

  • Patient Global Impression ‐ improvement (PGI‐I) at endpoint

  • Clinical Global Impression of improvement (CGI‐I) at endpoint

  • Fibromyalgia Impact Questionnaire (FIQ)

  • 36‐Item Short‐Form Health Survey (SF‐36)

  • Beck Depression Inventory‐II (BDI)

  • Widespread Pain Index and Symptom Severity in American College of Rheumatology Fibromyalgia Diagnostic Criteria 2010

  • Average Pain and Worst Pain Severity Score within 24‐hours in Patient Diary

  • BPI Pain Severity Items and Interference Items of the BPI‐Modified Short Form Score

Starting date

March 2012

Contact information

Eli Lilly and Company, Shionogi. Tel: 1‐877‐CTLILLY (1‐877‐285‐4559) or 1‐317‐615‐4559

Notes

NCT01552057

Data and analyses

Open in table viewer
Comparison 1. Duloxetine versus placebo in the treatment of painful diabetic neuropathy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.

1.1 Duloxetine 20 mg daily

1

213

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.98, 2.09]

1.2 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Random, 95% CI)

1.91 [1.26, 2.87]

1.3 Duloxetine 60 mg daily

4

908

Risk Ratio (M‐H, Random, 95% CI)

1.73 [1.44, 2.08]

1.4 Duloxetine 120 mg daily

4

870

Risk Ratio (M‐H, Random, 95% CI)

1.46 [1.08, 1.97]

1.5 All doses

5

1655

Risk Ratio (M‐H, Random, 95% CI)

1.53 [1.21, 1.92]

2 Mean improvement in pain at 12 weeks or less Show forest plot

5

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.

2.1 Duloxetine 20 mg daily

1

179

Mean Difference (IV, Fixed, 95% CI)

‐0.45 [‐1.05, 0.15]

2.2 Duloxetine 60 mg daily

4

722

Mean Difference (IV, Fixed, 95% CI)

‐0.96 [‐1.26, ‐0.65]

2.3 Duloxetine 120 mg daily

4

828

Mean Difference (IV, Fixed, 95% CI)

‐0.93 [‐1.21, ‐0.65]

3 Number of participants with ≥ 30% improvement in pain at 12 weeks or less Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 3 Number of participants with ≥ 30% improvement in pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 3 Number of participants with ≥ 30% improvement in pain at 12 weeks or less.

3.1 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

1.57 [1.18, 2.07]

3.2 Duloxetine 60 mg daily

4

799

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [1.33, 1.75]

3.3 Duloxetine 120 mg daily

3

659

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [1.21, 1.58]

3.4 All doses

4

1220

Risk Ratio (M‐H, Fixed, 95% CI)

1.45 [1.30, 1.63]

4 Mean improvement in SF‐36 Physical Subscore at 12 weeks or less Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 4 Mean improvement in SF‐36 Physical Subscore at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 4 Mean improvement in SF‐36 Physical Subscore at 12 weeks or less.

4.1 Duloxetine 20 mg daily

1

200

Mean Difference (IV, Random, 95% CI)

‐0.27 [‐2.42, 1.88]

4.2 Duloxetine 60 mg daily

3

514

Mean Difference (IV, Random, 95% CI)

2.65 [1.38, 3.92]

4.3 Duloxetine 120 mg daily

2

409

Mean Difference (IV, Random, 95% CI)

2.80 [1.04, 4.55]

5 Mean improvement in SF‐36 Mental Subscore at 12 weeks or less Show forest plot

3

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 5 Mean improvement in SF‐36 Mental Subscore at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 5 Mean improvement in SF‐36 Mental Subscore at 12 weeks or less.

5.1 Duloxetine 20 mg daily

1

200

Mean Difference (IV, Fixed, 95% CI)

1.11 [‐0.98, 3.20]

5.2 Duloxetine 60 mg daily

3

514

Mean Difference (IV, Fixed, 95% CI)

1.08 [‐0.32, 2.48]

5.3 Duloxetine 120 mg daily

2

409

Mean Difference (IV, Fixed, 95% CI)

2.23 [0.69, 3.77]

6 Mean improvement in SF‐36 Bodily Pain Subscore at 12 weeks or less Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.6

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 6 Mean improvement in SF‐36 Bodily Pain Subscore at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 6 Mean improvement in SF‐36 Bodily Pain Subscore at 12 weeks or less.

6.1 Duloxetine 20 mg daily

1

209

Mean Difference (IV, Fixed, 95% CI)

2.90 [‐2.37, 8.17]

6.2 Duloxetine 60 mg daily

2

421

Mean Difference (IV, Fixed, 95% CI)

5.58 [1.74, 9.42]

6.3 Duloxetine 120 mg daily

2

420

Mean Difference (IV, Fixed, 95% CI)

8.19 [4.33, 12.05]

7 Mean improvement in Patient Reported Global Impression of Improvement at 12 weeks or less Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.7

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 7 Mean improvement in Patient Reported Global Impression of Improvement at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 7 Mean improvement in Patient Reported Global Impression of Improvement at 12 weeks or less.

7.1 Duloxetine 20 mg daily

1

219

Mean Difference (IV, Random, 95% CI)

‐0.23 [‐0.56, 0.10]

7.2 Duloxetine 40 mg daily

1

252

Mean Difference (IV, Random, 95% CI)

‐0.65 [‐1.01, ‐0.29]

7.3 Duloxetine 60 mg daily

5

1018

Mean Difference (IV, Random, 95% CI)

‐0.60 [‐0.77, ‐0.44]

7.4 Duloxetine 120 mg daily

4

870

Mean Difference (IV, Random, 95% CI)

‐0.54 [‐0.73, ‐0.35]

8 Mean improvement in BPI Severity ‐ average pain at 12 weeks or less Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.8

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 8 Mean improvement in BPI Severity ‐ average pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 8 Mean improvement in BPI Severity ‐ average pain at 12 weeks or less.

8.1 Duloxetine 60 mg daily

2

433

Mean Difference (IV, Random, 95% CI)

‐0.97 [‐1.38, ‐0.57]

8.2 Duloxetine 120 mg daily

2

428

Mean Difference (IV, Random, 95% CI)

‐1.16 [‐1.91, ‐0.41]

9 Mean improvement in pain at rest (night pain) at 12 weeks or less Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.9

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 9 Mean improvement in pain at rest (night pain) at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 9 Mean improvement in pain at rest (night pain) at 12 weeks or less.

9.1 Duloxetine 20 mg daily

1

222

Mean Difference (IV, Random, 95% CI)

‐0.28 [‐0.90, 0.34]

9.2 Duloxetine 60 mg daily

3

664

Mean Difference (IV, Random, 95% CI)

‐0.92 [‐1.27, ‐0.57]

9.3 Duloxetine 120 mg daily

3

664

Mean Difference (IV, Random, 95% CI)

‐1.10 [‐1.45, ‐0.75]

Open in table viewer
Comparison 2. Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with ≥ 50% improvement in pain at 12 weeks or less Show forest plot

1

804

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [1.19, 1.80]

Analysis 2.1

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement in pain at 12 weeks or less.

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement in pain at 12 weeks or less.

2 Mean improvement in pain at 12 weeks or less Show forest plot

1

804

Mean Difference (IV, Fixed, 95% CI)

‐0.62 [‐0.92, ‐0.32]

Analysis 2.2

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.

3 Number improved ≥ 30% at 12 weeks or less Show forest plot

1

804

Risk Ratio (M‐H, Fixed, 95% CI)

1.42 [1.20, 1.68]

Analysis 2.3

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 3 Number improved ≥ 30% at 12 weeks or less.

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 3 Number improved ≥ 30% at 12 weeks or less.

Open in table viewer
Comparison 3. Duloxetine versus placebo in the treatment of fibromyalgia

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.1

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.

1.1 Duloxetine 20 mg daily

1

223

Risk Ratio (M‐H, Fixed, 95% CI)

1.39 [0.91, 2.14]

1.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.75, 1.35]

1.3 Duloxetine 60 mg daily

2

528

Risk Ratio (M‐H, Fixed, 95% CI)

1.57 [1.20, 2.06]

1.4 Duloxetine 120 mg daily

4

1234

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [1.40, 2.03]

1.5 All doses

5

1887

Risk Ratio (M‐H, Fixed, 95% CI)

1.50 [1.29, 1.75]

2 Number of participants with ≥ 50% improvement of pain at more than 12 weeks Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.2

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 2 Number of participants with ≥ 50% improvement of pain at more than 12 weeks.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 2 Number of participants with ≥ 50% improvement of pain at more than 12 weeks.

2.1 Duloxetine 60 mg daily

1

373

Risk Ratio (M‐H, Fixed, 95% CI)

1.58 [1.10, 2.27]

2.2 Duloxetine 120 mg daily

2

616

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [1.07, 1.79]

2.3 Duloxetine all doses

2

845

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [1.09, 1.79]

3 Number of participants with ≥ 30% improvement of pain at 12 weeks or less Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.3

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 3 Number of participants with ≥ 30% improvement of pain at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 3 Number of participants with ≥ 30% improvement of pain at 12 weeks or less.

3.1 Duloxetine 20 mg daily

1

223

Risk Ratio (M‐H, Fixed, 95% CI)

1.30 [0.94, 1.79]

3.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.89, 1.45]

3.3 Duloxetine 60 mg daily

2

528

Risk Ratio (M‐H, Fixed, 95% CI)

1.52 [1.24, 1.85]

3.4 Duloxetine 120 mg daily

3

1020

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [1.26, 1.69]

3.5 All doses

4

1673

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [1.22, 1.56]

4 Mean improvement in pain at 12 weeks or less Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 3.4

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 4 Mean improvement in pain at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 4 Mean improvement in pain at 12 weeks or less.

4.1 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Fixed, 95% CI)

‐0.31 [‐0.86, 0.24]

4.2 Duloxetine 120 mg daily

1

507

Mean Difference (IV, Fixed, 95% CI)

‐0.80 [‐1.35, ‐0.25]

5 Mean improvement in the SF‐36 mental component summary subscore Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 3.5

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 5 Mean improvement in the SF‐36 mental component summary subscore.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 5 Mean improvement in the SF‐36 mental component summary subscore.

5.1 Duloxetine 20 mg daily

1

223

Mean Difference (IV, Random, 95% CI)

0.81 [‐2.37, 3.99]

5.2 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Random, 95% CI)

2.69 [0.31, 5.07]

5.3 Duloxetine 60 mg daily

2

515

Mean Difference (IV, Random, 95% CI)

3.31 [0.59, 6.02]

5.4 Duloxetine 120 mg daily

5

1531

Mean Difference (IV, Random, 95% CI)

4.22 [2.43, 6.02]

6 Mean improvement in the SF‐36 physical component summary subscore Show forest plot

6

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 3.6

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 6 Mean improvement in the SF‐36 physical component summary subscore.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 6 Mean improvement in the SF‐36 physical component summary subscore.

6.1 Duloxetine 20 mg daily

1

223

Mean Difference (IV, Fixed, 95% CI)

0.81 [‐1.92, 3.54]

6.2 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Fixed, 95% CI)

0.84 [‐1.17, 2.85]

6.3 Duloxetine 60 mg daily

2

515

Mean Difference (IV, Fixed, 95% CI)

1.28 [‐0.33, 2.89]

6.4 Duloxetine 120 mg daily

5

1531

Mean Difference (IV, Fixed, 95% CI)

2.13 [0.95, 3.30]

7 Mean improvement in the SF‐36 Bodily Pain Subscore Show forest plot

4

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 3.7

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 7 Mean improvement in the SF‐36 Bodily Pain Subscore.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 7 Mean improvement in the SF‐36 Bodily Pain Subscore.

7.1 Duloxetine 60 mg daily

1

221

Mean Difference (IV, Fixed, 95% CI)

8.2 [3.20, 13.20]

7.2 Duloxetine 120 mg daily

4

1243

Mean Difference (IV, Fixed, 95% CI)

5.96 [3.76, 8.16]

8 Mean improvement in the Patient reported Global Impression of Change at completion of trial Show forest plot

4

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 3.8

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 8 Mean improvement in the Patient reported Global Impression of Change at completion of trial.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 8 Mean improvement in the Patient reported Global Impression of Change at completion of trial.

8.1 Duloxetine 20 mg daily

1

223

Mean Difference (IV, Fixed, 95% CI)

‐0.54 [‐0.96, ‐0.12]

8.2 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Fixed, 95% CI)

‐0.38 [‐0.71, ‐0.05]

8.3 Duloxetine 60 mg daily

2

519

Mean Difference (IV, Fixed, 95% CI)

‐0.45 [‐0.73, ‐0.18]

8.4 Duloxetine 120 mg daily

3

826

Mean Difference (IV, Fixed, 95% CI)

‐0.44 [‐0.66, ‐0.23]

Open in table viewer
Comparison 4. Duloxetine versus placebo for the treatment of pain in major depressive disorder

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with > 50% pain relief at 12 weeks or less Show forest plot

2

1023

Risk Ratio (M‐H, Fixed, 95% CI)

1.37 [1.19, 1.59]

Analysis 4.1

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 1 Number of participants with > 50% pain relief at 12 weeks or less.

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 1 Number of participants with > 50% pain relief at 12 weeks or less.

2 Participants with > 30% pain relief at 12 weeks or less Show forest plot

3

1359

Risk Ratio (M‐H, Fixed, 95% CI)

1.27 [1.15, 1.40]

Analysis 4.2

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 2 Participants with > 30% pain relief at 12 weeks or less.

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 2 Participants with > 30% pain relief at 12 weeks or less.

3 Mean improvement in pain at 12 weeks or less Show forest plot

3

1359

Mean Difference (IV, Fixed, 95% CI)

‐0.55 [‐0.75, ‐0.35]

Analysis 4.3

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 3 Mean improvement in pain at 12 weeks or less.

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 3 Mean improvement in pain at 12 weeks or less.

Open in table viewer
Comparison 5. Duloxetine versus placebo in the treatment of central neuropathic pain

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mean improvement in pain at 12 weeks or less Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

‐1.0 [‐2.05, 0.05]

Analysis 5.1

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 1 Mean improvement in pain at 12 weeks or less.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 1 Mean improvement in pain at 12 weeks or less.

2 Mean improvement in SF‐36 Physical Subscore Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

2.0 [‐12.72, 16.72]

Analysis 5.2

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 2 Mean improvement in SF‐36 Physical Subscore.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 2 Mean improvement in SF‐36 Physical Subscore.

3 Mean improvement in the SF‐36 Mental Subscore at 12 weeks Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

4.0 [‐6.75, 14.75]

Analysis 5.3

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 3 Mean improvement in the SF‐36 Mental Subscore at 12 weeks.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 3 Mean improvement in the SF‐36 Mental Subscore at 12 weeks.

4 Mean improvement in the SF‐36 Bodily Pain Subscore Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

8.0 [‐0.81, 16.81]

Analysis 5.4

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 4 Mean improvement in the SF‐36 Bodily Pain Subscore.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 4 Mean improvement in the SF‐36 Bodily Pain Subscore.

5 Number of participants improved on PGI‐I (better or very much better) Show forest plot

1

48

Risk Ratio (M‐H, Fixed, 95% CI)

2.75 [1.02, 7.44]

Analysis 5.5

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 5 Number of participants improved on PGI‐I (better or very much better).

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 5 Number of participants improved on PGI‐I (better or very much better).

Open in table viewer
Comparison 6. Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of participants with any adverse event Show forest plot

14

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 6.1

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 1 Proportion of participants with any adverse event.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 1 Proportion of participants with any adverse event.

1.1 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.25 [1.03, 1.52]

1.2 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.01, 1.31]

1.3 Duloxetine 60 mg daily

13

4521

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.10, 1.20]

1.4 Duloxetine 120 mg daily

3

688

Risk Ratio (M‐H, Fixed, 95% CI)

1.19 [1.09, 1.30]

1.5 Duloxetine all doses

14

5258

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.11, 1.20]

2 Nausea Show forest plot

13

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 6.2

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 2 Nausea.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 2 Nausea.

2.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Random, 95% CI)

1.45 [0.71, 3.00]

2.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Random, 95% CI)

5.43 [2.34, 12.58]

2.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Random, 95% CI)

6.55 [1.85, 23.17]

2.4 Duloxetine 60 mg daily

11

3642

Risk Ratio (M‐H, Random, 95% CI)

2.61 [2.14, 3.18]

2.5 Duloxetine 120 mg daily

4

787

Risk Ratio (M‐H, Random, 95% CI)

2.89 [2.06, 4.04]

3 Dry mouth Show forest plot

7

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 6.3

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 3 Dry mouth.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 3 Dry mouth.

3.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.30, 2.47]

3.2 Duloxetine 40 mg daily

0

0

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.3 Duloxetine 60 mg daily

6

2004

Risk Ratio (M‐H, Fixed, 95% CI)

2.63 [1.89, 3.67]

3.4 Duloxetine 120 mg daily

3

567

Risk Ratio (M‐H, Fixed, 95% CI)

3.40 [1.94, 5.96]

4 Dizziness Show forest plot

9

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 6.4

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 4 Dizziness.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 4 Dizziness.

4.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.33, 2.33]

4.2 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

5.89 [1.22, 28.58]

4.3 Duloxetine 60 mg daily

8

2257

Risk Ratio (M‐H, Fixed, 95% CI)

1.84 [1.35, 2.51]

4.4 Duloxetine 120 mg daily

4

787

Risk Ratio (M‐H, Fixed, 95% CI)

2.44 [1.55, 3.83]

5 Somnolence Show forest plot

10

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 6.5

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 5 Somnolence.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 5 Somnolence.

5.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.41, 2.43]

5.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

2.22 [0.70, 7.06]

5.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

2.25 [1.15, 4.38]

5.4 Duloxetine 60 mg daily

8

2678

Risk Ratio (M‐H, Fixed, 95% CI)

2.94 [2.17, 3.97]

5.5 Duloxetine 120 mg daily

4

787

Risk Ratio (M‐H, Fixed, 95% CI)

4.76 [2.93, 7.74]

6 Adverse event leading to cessation Show forest plot

17

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 6.6

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 6 Adverse event leading to cessation.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 6 Adverse event leading to cessation.

6.1 Duloxetine 20 mg daily

2

453

Risk Ratio (M‐H, Fixed, 95% CI)

1.37 [0.78, 2.39]

6.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.54 [0.69, 3.44]

6.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

1.96 [0.81, 4.77]

6.4 Duloxetine 60 mg daily

14

4837

Risk Ratio (M‐H, Fixed, 95% CI)

1.95 [1.60, 2.37]

6.5 Duloxetine 120 mg daily

7

1462

Risk Ratio (M‐H, Fixed, 95% CI)

2.30 [1.74, 3.04]

6.6 All doses

17

6285

Risk Ratio (M‐H, Fixed, 95% CI)

1.99 [1.67, 2.37]

7 Serious adverse event Show forest plot

16

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 6.7

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 7 Serious adverse event.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 7 Serious adverse event.

7.1 Duloxetine 20 mg daily

0

0

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.01, 8.02]

7.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

2.95 [0.50, 17.30]

7.4 Duloxetine 60 mg daily

14

4842

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.60, 1.32]

7.5 Duloxetine 120 mg daily

6

1257

Risk Ratio (M‐H, Fixed, 95% CI)

0.59 [0.25, 1.35]

7.6 All doses

14

4976

Risk Ratio (M‐H, Fixed, 95% CI)

0.81 [0.53, 1.25]

Methodological quality summary: review authors' judgements about each methodological quality item for each included study. Green = low risk of bias; yellow = unclear risk of bias; red = high risk of bias
Figuras y tablas -
Figure 1

Methodological quality summary: review authors' judgements about each methodological quality item for each included study. Green = low risk of bias; yellow = unclear risk of bias; red = high risk of bias

Duloxetine versus placebo in the treatment of painful neuropathy: Number of patients with >50% improvement of pain at <12 weeks.
Figuras y tablas -
Figure 2

Duloxetine versus placebo in the treatment of painful neuropathy: Number of patients with >50% improvement of pain at <12 weeks.

Duloxetine versus placebo in the treatment of pain: Mean improvement in pain at 12 weeks.
Figuras y tablas -
Figure 3

Duloxetine versus placebo in the treatment of pain: Mean improvement in pain at 12 weeks.

Duloxetine versus placebo in the treatment of pain: Number of patients with >30% improvement in pain at <12 weeks.
Figuras y tablas -
Figure 4

Duloxetine versus placebo in the treatment of pain: Number of patients with >30% improvement in pain at <12 weeks.

Duloxetine versus placebo in the treatment of pain: Patient reported global impression of change.
Figuras y tablas -
Figure 5

Duloxetine versus placebo in the treatment of pain: Patient reported global impression of change.

Duloxetine versus placebo in the treatment of pain: BPI severity ‐ average pain.
Figuras y tablas -
Figure 6

Duloxetine versus placebo in the treatment of pain: BPI severity ‐ average pain.

Trial sequential analysis of duloxetine versus placebo in the treatment of painful neuropathy ‐ 50% or more reduction in pain at 8‐12 weeks with at least 8 weeks of treatment
Figuras y tablas -
Figure 7

Trial sequential analysis of duloxetine versus placebo in the treatment of painful neuropathy ‐ 50% or more reduction in pain at 8‐12 weeks with at least 8 weeks of treatment

Duloxetine versus placebo in the treatment of fibromyalgia: >30% improvement <12 weeks.
Figuras y tablas -
Figure 8

Duloxetine versus placebo in the treatment of fibromyalgia: >30% improvement <12 weeks.

Duloxetine versus placebo in the treatment of fibromyalgia: SF‐36 bodily pain.
Figuras y tablas -
Figure 9

Duloxetine versus placebo in the treatment of fibromyalgia: SF‐36 bodily pain.

Trial sequential analysis of duloxetine 60 mg versus placebo for the 50% reduction in pain in fibromyalgia with at least 8 weeks treatment at 8‐12 weeks
Figuras y tablas -
Figure 10

Trial sequential analysis of duloxetine 60 mg versus placebo for the 50% reduction in pain in fibromyalgia with at least 8 weeks treatment at 8‐12 weeks

Trial Sequential Analysis of duloxetine 60 mg versus placebo in the treatment of painful physical symptoms in depression at less than 12 weeks with at least eight weeks of treatment
Figuras y tablas -
Figure 11

Trial Sequential Analysis of duloxetine 60 mg versus placebo in the treatment of painful physical symptoms in depression at less than 12 weeks with at least eight weeks of treatment

Adverse events leading to cessation of treatment.
Figuras y tablas -
Figure 12

Adverse events leading to cessation of treatment.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.
Figuras y tablas -
Analysis 1.1

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 1.2

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 3 Number of participants with ≥ 30% improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 1.3

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 3 Number of participants with ≥ 30% improvement in pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 4 Mean improvement in SF‐36 Physical Subscore at 12 weeks or less.
Figuras y tablas -
Analysis 1.4

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 4 Mean improvement in SF‐36 Physical Subscore at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 5 Mean improvement in SF‐36 Mental Subscore at 12 weeks or less.
Figuras y tablas -
Analysis 1.5

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 5 Mean improvement in SF‐36 Mental Subscore at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 6 Mean improvement in SF‐36 Bodily Pain Subscore at 12 weeks or less.
Figuras y tablas -
Analysis 1.6

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 6 Mean improvement in SF‐36 Bodily Pain Subscore at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 7 Mean improvement in Patient Reported Global Impression of Improvement at 12 weeks or less.
Figuras y tablas -
Analysis 1.7

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 7 Mean improvement in Patient Reported Global Impression of Improvement at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 8 Mean improvement in BPI Severity ‐ average pain at 12 weeks or less.
Figuras y tablas -
Analysis 1.8

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 8 Mean improvement in BPI Severity ‐ average pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 9 Mean improvement in pain at rest (night pain) at 12 weeks or less.
Figuras y tablas -
Analysis 1.9

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 9 Mean improvement in pain at rest (night pain) at 12 weeks or less.

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 2.1

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement in pain at 12 weeks or less.

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 2.2

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 3 Number improved ≥ 30% at 12 weeks or less.
Figuras y tablas -
Analysis 2.3

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 3 Number improved ≥ 30% at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.
Figuras y tablas -
Analysis 3.1

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 2 Number of participants with ≥ 50% improvement of pain at more than 12 weeks.
Figuras y tablas -
Analysis 3.2

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 2 Number of participants with ≥ 50% improvement of pain at more than 12 weeks.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 3 Number of participants with ≥ 30% improvement of pain at 12 weeks or less.
Figuras y tablas -
Analysis 3.3

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 3 Number of participants with ≥ 30% improvement of pain at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 4 Mean improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 3.4

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 4 Mean improvement in pain at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 5 Mean improvement in the SF‐36 mental component summary subscore.
Figuras y tablas -
Analysis 3.5

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 5 Mean improvement in the SF‐36 mental component summary subscore.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 6 Mean improvement in the SF‐36 physical component summary subscore.
Figuras y tablas -
Analysis 3.6

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 6 Mean improvement in the SF‐36 physical component summary subscore.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 7 Mean improvement in the SF‐36 Bodily Pain Subscore.
Figuras y tablas -
Analysis 3.7

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 7 Mean improvement in the SF‐36 Bodily Pain Subscore.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 8 Mean improvement in the Patient reported Global Impression of Change at completion of trial.
Figuras y tablas -
Analysis 3.8

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 8 Mean improvement in the Patient reported Global Impression of Change at completion of trial.

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 1 Number of participants with > 50% pain relief at 12 weeks or less.
Figuras y tablas -
Analysis 4.1

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 1 Number of participants with > 50% pain relief at 12 weeks or less.

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 2 Participants with > 30% pain relief at 12 weeks or less.
Figuras y tablas -
Analysis 4.2

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 2 Participants with > 30% pain relief at 12 weeks or less.

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 3 Mean improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 4.3

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 3 Mean improvement in pain at 12 weeks or less.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 1 Mean improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 5.1

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 1 Mean improvement in pain at 12 weeks or less.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 2 Mean improvement in SF‐36 Physical Subscore.
Figuras y tablas -
Analysis 5.2

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 2 Mean improvement in SF‐36 Physical Subscore.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 3 Mean improvement in the SF‐36 Mental Subscore at 12 weeks.
Figuras y tablas -
Analysis 5.3

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 3 Mean improvement in the SF‐36 Mental Subscore at 12 weeks.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 4 Mean improvement in the SF‐36 Bodily Pain Subscore.
Figuras y tablas -
Analysis 5.4

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 4 Mean improvement in the SF‐36 Bodily Pain Subscore.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 5 Number of participants improved on PGI‐I (better or very much better).
Figuras y tablas -
Analysis 5.5

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 5 Number of participants improved on PGI‐I (better or very much better).

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 1 Proportion of participants with any adverse event.
Figuras y tablas -
Analysis 6.1

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 1 Proportion of participants with any adverse event.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 2 Nausea.
Figuras y tablas -
Analysis 6.2

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 2 Nausea.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 3 Dry mouth.
Figuras y tablas -
Analysis 6.3

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 3 Dry mouth.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 4 Dizziness.
Figuras y tablas -
Analysis 6.4

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 4 Dizziness.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 5 Somnolence.
Figuras y tablas -
Analysis 6.5

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 5 Somnolence.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 6 Adverse event leading to cessation.
Figuras y tablas -
Analysis 6.6

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 6 Adverse event leading to cessation.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 7 Serious adverse event.
Figuras y tablas -
Analysis 6.7

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 7 Serious adverse event.

Summary of findings for the main comparison. Duloxetine for the treatment of painful diabetic neuropathy

Duloxetine for painful diabetic neuropathy

Patient or population: patients with painful neuropathy or chronic pain from diabetic peripheral neuropathy
Settings: primary and secondary care
Intervention: duloxetine

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Duloxetine

Number of patients with ≥ 50% improvement of pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert score

Follow‐up: 8 to 12 weeks

257 per 1000

445 per 1000
(370 to 535)

RR 1.73
(1.44 to 2.08)

908
(4 studies)

⊕⊕⊕⊝
moderate1

NNTB for ≥ 50% reduction in pain at 60 mg daily: 5 (95% CI 4 to 7)

Mean improvement in pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert score

Scale from: 0 to 10
Follow‐up: 8 to 12 weeks

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 60 mg daily in the control groups was
‐1.65 units

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 60 mg daily in the intervention groups was
0.96 lower
(1.26 to 0.65 lower)

722
(4 studies)

⊕⊕⊕⊝
moderate2

Number of patients with ≥ 30% improvement in pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert scale

Follow‐up: 8 to 12 weeks

411 per 1000

629 per 1000
(547 to 719)

RR 1.53
(1.33 to 1.75)

799
(4 studies)

⊕⊕⊕⊝
moderate1

NNTB for ≥ 30% reduction in pain at 60 mg duloxetine daily: 5 (95% CI 3 to 7)

Mean improvement in Patient Reported Global Impression of Change at 12 weeks or less

Duloxetine 60 mg daily
VAS

Scale from: 0 to 10
Follow‐up: 8 to 12 weeks

The mean mean improvement in patient reported global impression of improvement change at 12 weeks or less ‐ duloxetine 60 mg daily in the control groups was
‐3.06 units

The mean mean improvement in Patient Reported Global Impression of Improvement Change at 12 weeks or less ‐ duloxetine 60 mg daily in the intervention groups was
0.6 lower
(0.77 to 0.44 lower)

1018
(5 studies)

⊕⊕⊕⊝
moderate3

Adverse event leading to cessation

All neuropathic pain indications

Duloxetine 60 mg daily

56 per 1000

109 per 1000
(90 to 133)

RR 1.95
(1.6 to 2.37)

4837
(14 studies)

⊕⊕⊝⊝
low4

NNTH for duloxetine 60 mg daily, all indications, and all adverse effects leading to cessation: 18 (95% CI 13 to 30)

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; NNTB: number needed to treat for an additional beneficial outcome; NNTH: number needed to treat for an additional harmful outcome

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

1 Four trials, all company sponsored and performed but all trials pre‐registered on ClinicalTrials.gov have been published. No publication bias detected.
2 Two of four studies by company. Effect in Rowbotham nonsignificant, contributing some heterogeneity.
3 Five studies but wide CIs in the independent studies.
4 Variable quality of adverse event collection.

Figuras y tablas -
Summary of findings for the main comparison. Duloxetine for the treatment of painful diabetic neuropathy
Summary of findings 2. Duloxetine for the treatment of the chronic pain of fibromyalgia

Duloxetine for the chronic pain of fibromyalgia

Patient or population: patients with the chronic pain of fibromyalgia
Settings:
Intervention: duloxetine

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Duloxetine

Number with ≥ 50% improvement of pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert scale
Follow‐up: 8 to 12 weeks

233 per 1000

366 per 1000
(280 to 480)

RR 1.57
(1.2 to 2.06)

528
(2 studies)

⊕⊕⊝⊝
low1,2

NNTB for ≥ 50% improvement of pain at duloxetine 60 mg daily: 8 (95% CI 4 to 21)

Number with ≥ 30% improvement of pain at 12 weeks or less

Duloxetine 60 mg daily

Follow‐up: 8 to 12 weeks

347 per 1000

527 per 1000
(430 to 642)

RR 1.52
(1.24 to 1.85)

528
(2 studies)

⊕⊕⊝⊝
low1,2

NNTB for ≥ 30% improvement of pain at duloxetine 60 mg daily: NNT 6 (95% CI 3 to 12)

Mean improvement in the Patient Reported Global Impression of Change at completion of trial

Duloxetine 60 mg daily
VAS

Scale from: 0 to 10
Follow‐up: 12 weeks

The mean mean improvement in the patient reported global impression of change at completion of trial ‐ duloxetine 60 mg daily in the control groups was
3.52 units

The mean mean improvement in the patient reported global impression of change at completion of trial ‐ duloxetine 60 mg daily in the intervention groups was
0.45 lower
(0.73 to 0.18 lower)

519
(2 studies)

⊕⊕⊝⊝
low1,2

Mean improvement in pain at 12 weeks or less

Duloxetine 120 mg daily

LikertScale from: 0 to 10
Follow‐up: 12 weeks

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 120 mg daily in the control groups was
‐1.5

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 120 mg daily in the intervention groups was
0.8 lower
(1.35 to 0.25 lower)

507
(1 study)

⊕⊕⊕⊝
moderate1

Adverse events

See comment

See comment

See comment

See comment

See pooled adverse events in 'Summary of findings' table 1

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; NNTB: number needed to treat for an additional beneficial outcome

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

1 Substantial dropouts from all trials inform the outcomes.
2 Mostly female in some trials, all female in others.

Figuras y tablas -
Summary of findings 2. Duloxetine for the treatment of the chronic pain of fibromyalgia
Summary of findings 3. Duloxetine for the treatment of pain in major depressive disorder

Duloxetine for pain in major depressive disorder

Patient or population: patients with pain in major depressive disorder
Settings:
Intervention: duloxetine

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Duloxetine

Number with ≥ 50% pain relief at 12 weeks or less
Follow‐up: 12 weeks

360 per 1000

493 per 1000
(428 to 572)

RR 1.37
(1.19 to 1.59)

1023
(2 studies)

⊕⊕⊕⊝
moderate1

NNTB for ≥ 50% pain relief at < 12 weeks 60 mg duloxetine daily: 8 (95% CI 5 to 14)

Number with ≥ 30% pain relief at 12 weeks or less

467 per 1000

593 per 1000
(537 to 654)

RR 1.27
(1.15 to 1.4)

1359
(3 studies)

⊕⊕⊝⊝
low1,2

NNTB for ≥ 30% pain relief at < 12 weeks 60 mg duloxetine: 8 (95% CI 4‐ to 14)

Mean improvement in pain at 12 weeks or less
Visual analogue scale. Scale from: 0 to 10.
Follow‐up: 12 weeks

The mean mean improvement in pain at 12 weeks or less in the control groups was
1.23

The mean mean improvement in pain at 12 weeks or less in the intervention groups was
0.55 lower
(0.75 to 0.35 lower)

1359
(3 studies)

⊕⊕⊝⊝
low1,2

Mean improvement in Patient Reported Global Impression of Change at 12 weeks or less

See comment

See comment

Not estimable

See comment

Outcome not measured

Adverse events

See comment

See comment

Not estimable

See comment

See pooled adverse events in 'Summary of findings' table 1

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; NNTB: number needed to treat for an additional beneficial outcome

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

1 Mixed causes for pain, not necessarily neuropathic.
2 Substantial dropouts partially accounted for by last observation carried forward and statistical manipulation.

Figuras y tablas -
Summary of findings 3. Duloxetine for the treatment of pain in major depressive disorder
Comparison 1. Duloxetine versus placebo in the treatment of painful diabetic neuropathy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Duloxetine 20 mg daily

1

213

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.98, 2.09]

1.2 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Random, 95% CI)

1.91 [1.26, 2.87]

1.3 Duloxetine 60 mg daily

4

908

Risk Ratio (M‐H, Random, 95% CI)

1.73 [1.44, 2.08]

1.4 Duloxetine 120 mg daily

4

870

Risk Ratio (M‐H, Random, 95% CI)

1.46 [1.08, 1.97]

1.5 All doses

5

1655

Risk Ratio (M‐H, Random, 95% CI)

1.53 [1.21, 1.92]

2 Mean improvement in pain at 12 weeks or less Show forest plot

5

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

2.1 Duloxetine 20 mg daily

1

179

Mean Difference (IV, Fixed, 95% CI)

‐0.45 [‐1.05, 0.15]

2.2 Duloxetine 60 mg daily

4

722

Mean Difference (IV, Fixed, 95% CI)

‐0.96 [‐1.26, ‐0.65]

2.3 Duloxetine 120 mg daily

4

828

Mean Difference (IV, Fixed, 95% CI)

‐0.93 [‐1.21, ‐0.65]

3 Number of participants with ≥ 30% improvement in pain at 12 weeks or less Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

1.57 [1.18, 2.07]

3.2 Duloxetine 60 mg daily

4

799

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [1.33, 1.75]

3.3 Duloxetine 120 mg daily

3

659

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [1.21, 1.58]

3.4 All doses

4

1220

Risk Ratio (M‐H, Fixed, 95% CI)

1.45 [1.30, 1.63]

4 Mean improvement in SF‐36 Physical Subscore at 12 weeks or less Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 Duloxetine 20 mg daily

1

200

Mean Difference (IV, Random, 95% CI)

‐0.27 [‐2.42, 1.88]

4.2 Duloxetine 60 mg daily

3

514

Mean Difference (IV, Random, 95% CI)

2.65 [1.38, 3.92]

4.3 Duloxetine 120 mg daily

2

409

Mean Difference (IV, Random, 95% CI)

2.80 [1.04, 4.55]

5 Mean improvement in SF‐36 Mental Subscore at 12 weeks or less Show forest plot

3

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

5.1 Duloxetine 20 mg daily

1

200

Mean Difference (IV, Fixed, 95% CI)

1.11 [‐0.98, 3.20]

5.2 Duloxetine 60 mg daily

3

514

Mean Difference (IV, Fixed, 95% CI)

1.08 [‐0.32, 2.48]

5.3 Duloxetine 120 mg daily

2

409

Mean Difference (IV, Fixed, 95% CI)

2.23 [0.69, 3.77]

6 Mean improvement in SF‐36 Bodily Pain Subscore at 12 weeks or less Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

6.1 Duloxetine 20 mg daily

1

209

Mean Difference (IV, Fixed, 95% CI)

2.90 [‐2.37, 8.17]

6.2 Duloxetine 60 mg daily

2

421

Mean Difference (IV, Fixed, 95% CI)

5.58 [1.74, 9.42]

6.3 Duloxetine 120 mg daily

2

420

Mean Difference (IV, Fixed, 95% CI)

8.19 [4.33, 12.05]

7 Mean improvement in Patient Reported Global Impression of Improvement at 12 weeks or less Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

7.1 Duloxetine 20 mg daily

1

219

Mean Difference (IV, Random, 95% CI)

‐0.23 [‐0.56, 0.10]

7.2 Duloxetine 40 mg daily

1

252

Mean Difference (IV, Random, 95% CI)

‐0.65 [‐1.01, ‐0.29]

7.3 Duloxetine 60 mg daily

5

1018

Mean Difference (IV, Random, 95% CI)

‐0.60 [‐0.77, ‐0.44]

7.4 Duloxetine 120 mg daily

4

870

Mean Difference (IV, Random, 95% CI)

‐0.54 [‐0.73, ‐0.35]

8 Mean improvement in BPI Severity ‐ average pain at 12 weeks or less Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

8.1 Duloxetine 60 mg daily

2

433

Mean Difference (IV, Random, 95% CI)

‐0.97 [‐1.38, ‐0.57]

8.2 Duloxetine 120 mg daily

2

428

Mean Difference (IV, Random, 95% CI)

‐1.16 [‐1.91, ‐0.41]

9 Mean improvement in pain at rest (night pain) at 12 weeks or less Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

9.1 Duloxetine 20 mg daily

1

222

Mean Difference (IV, Random, 95% CI)

‐0.28 [‐0.90, 0.34]

9.2 Duloxetine 60 mg daily

3

664

Mean Difference (IV, Random, 95% CI)

‐0.92 [‐1.27, ‐0.57]

9.3 Duloxetine 120 mg daily

3

664

Mean Difference (IV, Random, 95% CI)

‐1.10 [‐1.45, ‐0.75]

Figuras y tablas -
Comparison 1. Duloxetine versus placebo in the treatment of painful diabetic neuropathy
Comparison 2. Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with ≥ 50% improvement in pain at 12 weeks or less Show forest plot

1

804

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [1.19, 1.80]

2 Mean improvement in pain at 12 weeks or less Show forest plot

1

804

Mean Difference (IV, Fixed, 95% CI)

‐0.62 [‐0.92, ‐0.32]

3 Number improved ≥ 30% at 12 weeks or less Show forest plot

1

804

Risk Ratio (M‐H, Fixed, 95% CI)

1.42 [1.20, 1.68]

Figuras y tablas -
Comparison 2. Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy
Comparison 3. Duloxetine versus placebo in the treatment of fibromyalgia

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

1.1 Duloxetine 20 mg daily

1

223

Risk Ratio (M‐H, Fixed, 95% CI)

1.39 [0.91, 2.14]

1.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.75, 1.35]

1.3 Duloxetine 60 mg daily

2

528

Risk Ratio (M‐H, Fixed, 95% CI)

1.57 [1.20, 2.06]

1.4 Duloxetine 120 mg daily

4

1234

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [1.40, 2.03]

1.5 All doses

5

1887

Risk Ratio (M‐H, Fixed, 95% CI)

1.50 [1.29, 1.75]

2 Number of participants with ≥ 50% improvement of pain at more than 12 weeks Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2.1 Duloxetine 60 mg daily

1

373

Risk Ratio (M‐H, Fixed, 95% CI)

1.58 [1.10, 2.27]

2.2 Duloxetine 120 mg daily

2

616

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [1.07, 1.79]

2.3 Duloxetine all doses

2

845

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [1.09, 1.79]

3 Number of participants with ≥ 30% improvement of pain at 12 weeks or less Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Duloxetine 20 mg daily

1

223

Risk Ratio (M‐H, Fixed, 95% CI)

1.30 [0.94, 1.79]

3.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.89, 1.45]

3.3 Duloxetine 60 mg daily

2

528

Risk Ratio (M‐H, Fixed, 95% CI)

1.52 [1.24, 1.85]

3.4 Duloxetine 120 mg daily

3

1020

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [1.26, 1.69]

3.5 All doses

4

1673

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [1.22, 1.56]

4 Mean improvement in pain at 12 weeks or less Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

4.1 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Fixed, 95% CI)

‐0.31 [‐0.86, 0.24]

4.2 Duloxetine 120 mg daily

1

507

Mean Difference (IV, Fixed, 95% CI)

‐0.80 [‐1.35, ‐0.25]

5 Mean improvement in the SF‐36 mental component summary subscore Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

5.1 Duloxetine 20 mg daily

1

223

Mean Difference (IV, Random, 95% CI)

0.81 [‐2.37, 3.99]

5.2 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Random, 95% CI)

2.69 [0.31, 5.07]

5.3 Duloxetine 60 mg daily

2

515

Mean Difference (IV, Random, 95% CI)

3.31 [0.59, 6.02]

5.4 Duloxetine 120 mg daily

5

1531

Mean Difference (IV, Random, 95% CI)

4.22 [2.43, 6.02]

6 Mean improvement in the SF‐36 physical component summary subscore Show forest plot

6

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

6.1 Duloxetine 20 mg daily

1

223

Mean Difference (IV, Fixed, 95% CI)

0.81 [‐1.92, 3.54]

6.2 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Fixed, 95% CI)

0.84 [‐1.17, 2.85]

6.3 Duloxetine 60 mg daily

2

515

Mean Difference (IV, Fixed, 95% CI)

1.28 [‐0.33, 2.89]

6.4 Duloxetine 120 mg daily

5

1531

Mean Difference (IV, Fixed, 95% CI)

2.13 [0.95, 3.30]

7 Mean improvement in the SF‐36 Bodily Pain Subscore Show forest plot

4

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

7.1 Duloxetine 60 mg daily

1

221

Mean Difference (IV, Fixed, 95% CI)

8.2 [3.20, 13.20]

7.2 Duloxetine 120 mg daily

4

1243

Mean Difference (IV, Fixed, 95% CI)

5.96 [3.76, 8.16]

8 Mean improvement in the Patient reported Global Impression of Change at completion of trial Show forest plot

4

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

8.1 Duloxetine 20 mg daily

1

223

Mean Difference (IV, Fixed, 95% CI)

‐0.54 [‐0.96, ‐0.12]

8.2 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Fixed, 95% CI)

‐0.38 [‐0.71, ‐0.05]

8.3 Duloxetine 60 mg daily

2

519

Mean Difference (IV, Fixed, 95% CI)

‐0.45 [‐0.73, ‐0.18]

8.4 Duloxetine 120 mg daily

3

826

Mean Difference (IV, Fixed, 95% CI)

‐0.44 [‐0.66, ‐0.23]

Figuras y tablas -
Comparison 3. Duloxetine versus placebo in the treatment of fibromyalgia
Comparison 4. Duloxetine versus placebo for the treatment of pain in major depressive disorder

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with > 50% pain relief at 12 weeks or less Show forest plot

2

1023

Risk Ratio (M‐H, Fixed, 95% CI)

1.37 [1.19, 1.59]

2 Participants with > 30% pain relief at 12 weeks or less Show forest plot

3

1359

Risk Ratio (M‐H, Fixed, 95% CI)

1.27 [1.15, 1.40]

3 Mean improvement in pain at 12 weeks or less Show forest plot

3

1359

Mean Difference (IV, Fixed, 95% CI)

‐0.55 [‐0.75, ‐0.35]

Figuras y tablas -
Comparison 4. Duloxetine versus placebo for the treatment of pain in major depressive disorder
Comparison 5. Duloxetine versus placebo in the treatment of central neuropathic pain

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mean improvement in pain at 12 weeks or less Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

‐1.0 [‐2.05, 0.05]

2 Mean improvement in SF‐36 Physical Subscore Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

2.0 [‐12.72, 16.72]

3 Mean improvement in the SF‐36 Mental Subscore at 12 weeks Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

4.0 [‐6.75, 14.75]

4 Mean improvement in the SF‐36 Bodily Pain Subscore Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

8.0 [‐0.81, 16.81]

5 Number of participants improved on PGI‐I (better or very much better) Show forest plot

1

48

Risk Ratio (M‐H, Fixed, 95% CI)

2.75 [1.02, 7.44]

Figuras y tablas -
Comparison 5. Duloxetine versus placebo in the treatment of central neuropathic pain
Comparison 6. Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of participants with any adverse event Show forest plot

14

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

1.1 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.25 [1.03, 1.52]

1.2 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.01, 1.31]

1.3 Duloxetine 60 mg daily

13

4521

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.10, 1.20]

1.4 Duloxetine 120 mg daily

3

688

Risk Ratio (M‐H, Fixed, 95% CI)

1.19 [1.09, 1.30]

1.5 Duloxetine all doses

14

5258

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.11, 1.20]

2 Nausea Show forest plot

13

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Random, 95% CI)

1.45 [0.71, 3.00]

2.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Random, 95% CI)

5.43 [2.34, 12.58]

2.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Random, 95% CI)

6.55 [1.85, 23.17]

2.4 Duloxetine 60 mg daily

11

3642

Risk Ratio (M‐H, Random, 95% CI)

2.61 [2.14, 3.18]

2.5 Duloxetine 120 mg daily

4

787

Risk Ratio (M‐H, Random, 95% CI)

2.89 [2.06, 4.04]

3 Dry mouth Show forest plot

7

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.30, 2.47]

3.2 Duloxetine 40 mg daily

0

0

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.3 Duloxetine 60 mg daily

6

2004

Risk Ratio (M‐H, Fixed, 95% CI)

2.63 [1.89, 3.67]

3.4 Duloxetine 120 mg daily

3

567

Risk Ratio (M‐H, Fixed, 95% CI)

3.40 [1.94, 5.96]

4 Dizziness Show forest plot

9

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

4.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.33, 2.33]

4.2 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

5.89 [1.22, 28.58]

4.3 Duloxetine 60 mg daily

8

2257

Risk Ratio (M‐H, Fixed, 95% CI)

1.84 [1.35, 2.51]

4.4 Duloxetine 120 mg daily

4

787

Risk Ratio (M‐H, Fixed, 95% CI)

2.44 [1.55, 3.83]

5 Somnolence Show forest plot

10

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

5.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.41, 2.43]

5.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

2.22 [0.70, 7.06]

5.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

2.25 [1.15, 4.38]

5.4 Duloxetine 60 mg daily

8

2678

Risk Ratio (M‐H, Fixed, 95% CI)

2.94 [2.17, 3.97]

5.5 Duloxetine 120 mg daily

4

787

Risk Ratio (M‐H, Fixed, 95% CI)

4.76 [2.93, 7.74]

6 Adverse event leading to cessation Show forest plot

17

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

6.1 Duloxetine 20 mg daily

2

453

Risk Ratio (M‐H, Fixed, 95% CI)

1.37 [0.78, 2.39]

6.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.54 [0.69, 3.44]

6.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

1.96 [0.81, 4.77]

6.4 Duloxetine 60 mg daily

14

4837

Risk Ratio (M‐H, Fixed, 95% CI)

1.95 [1.60, 2.37]

6.5 Duloxetine 120 mg daily

7

1462

Risk Ratio (M‐H, Fixed, 95% CI)

2.30 [1.74, 3.04]

6.6 All doses

17

6285

Risk Ratio (M‐H, Fixed, 95% CI)

1.99 [1.67, 2.37]

7 Serious adverse event Show forest plot

16

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

7.1 Duloxetine 20 mg daily

0

0

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.01, 8.02]

7.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

2.95 [0.50, 17.30]

7.4 Duloxetine 60 mg daily

14

4842

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.60, 1.32]

7.5 Duloxetine 120 mg daily

6

1257

Risk Ratio (M‐H, Fixed, 95% CI)

0.59 [0.25, 1.35]

7.6 All doses

14

4976

Risk Ratio (M‐H, Fixed, 95% CI)

0.81 [0.53, 1.25]

Figuras y tablas -
Comparison 6. Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia