Scolaris Content Display Scolaris Content Display

Serotonin and noradrenaline reuptake inhibitors (SNRIs) for fibromyalgia

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Arnold 2004 {published and unpublished data}

Arnold LM, Lu Y, Crofford LJ, Wohlreich M, Detke MJ, Iyengar S et al. 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. CENTRAL

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. CENTRAL

Arnold 2010a {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. Journal of Rheumatology 2010;37(12):2578-86. CENTRAL
Mohs R, Mease P, Arnold LM, Wang F, Ahl J, Gaynor PJ et al. The effect of duloxetine treatment on cognition in patients with fibromyalgia. Psychosomatic Medicine 2012;74:628-34. CENTRAL

Arnold 2010b {published data only}

Arnold LM, Gendreau RM, Palmer RH, Gendreau JF, Wang Y. Efficacy and safety of milnacipran 100 mg/day in patients with fibromyalgia: results of a randomized, double-blind, placebo-controlled trial. Arthritis and Rheumatism 2010;62(9):2745-56. CENTRAL

Arnold 2012a {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;28:775-81. CENTRAL

Bateman 2013 {published data only}

Bateman L, Palmer RH, Trugman JM, Lin Y. Results of switching to milnacipran in fibromyalgia patients with an inadequate response to duloxetine: a phase IV pilot study. Journal of Pain Research 2013;6:311-8. CENTRAL

Branco 2010 {published data only}

Branco JC, Zachrisson O, Perrot S, Mainguy Y, Multinational Coordinator Study Group. A European multicenter randomized double-blind placebo-controlled monotherapy clinical trial of milnacipran in treatment of fibromyalgia. Journal of Rheumatology 2010;37(4):851-9. CENTRAL

Chappell 2009a {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. CENTRAL

Clauw 2008 {published data only}

Clauw DJ, Mease P, Palmer RH, Gendreau RM, Wang Y. Milnacipran for the treatment of fibromyalgia in adults: a 15-week, multicenter, randomized, double-blind, placebo-controlled, multiple-dose clinical trial. Clinical Therapeutics 2008;30(11):1988-2004. CENTRAL

Clauw 2013 {published data only}

Clauw DJ, Mease PJ, Palmer RH, Trugman JM, Wang Y. Continuing efficacy of milnacipran following long-term treatment in fibromyalgia: a randomized trial. Arthritis Research & Therapy 2013;15:R88. CENTRAL
Mease PJ, Clauw DJ, Trugman JM, Palmer RH, Wang Y. Efficacy of long-term milnacipran treatment in patients meeting different thresholds of clinically relevant pain relief: subgroup analysis of a randomized, double-blind, placebo-controlled withdrawal study. Journal of Pain Research 2014;7:679-87. CENTRAL

Leombruni 2015 {published data only}

Leombruni P, Miniotti M, Colonna F, Sica C, Castelli L, Bruzzone M et al. A randomised controlled trial comparing duloxetine and acetyl L-carnitine in fibromyalgic patients: preliminary data. Clinical and Experimental Rheumatology 2015;33:S82-5. CENTRAL

Matthey 2013 {published data only}

Matthey A, Cedraschi C, Piguet V, Besson M, Chabert J, Daali Y et al. Dual reuptake inhibitor milnacipran and spinal pain pathways in fibromyalgia patients: a randomized, double-blind, placebo-controlled trial. Pain Physician 2013;16:E553-62. CENTRAL

Mease 2009b {published data only}

Mease PJ, Clauw DJ, Gendreau RM, Rao SG, Kranzler J, Chen W et al. The efficacy and safety of milnacipran for treatment of fibromyalgia. a randomized, double-blind, placebo-controlled trial. Journal of Rheumatology 2009;36(2):398-409. CENTRAL

Murakami 2015 {published data only}

Murakami M, Osada K, Mizuno H, Ochiai T, Nishioka K. A randomized, double-blind, placebo-controlled phase III trial of duloxetine in Japanese fibromyalgia patients. Arthritis Research & Therapy 2015;17:224. CENTRAL

NCT00697787 {published data only}

NCT00696787. A study evaluating desvenlafaxine sustained release (DVS SR) in adult female outpatients with fibromyalgia. clinicaltrials.gov/ct2/results?term=NCT00696787&Search=Search First posted: June 13, 2008. CENTRAL

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 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. CENTRAL

Staud 2015 {published data only}

Staud R, Lucas YE, Price DD, Robinson ME. Dual reuptake inhibitor milnacipran and spinal pain pathways in fibromyalgia patients: a randomized, double-blind, placebo-controlled trial. Journal of Pain 2015;16:750-9. CENTRAL

Vitton 2004 {published data only}

Gendreau RM, Thorn MD, Gendreau JF, Kranzler JD, Ribeiro S, Gracely RH et al. Efficacy of milnacipran in patients with fibromyalgia. Journal of Rheumatology 2005;32(10):1975-85. CENTRAL
Vitton O, Gendreau M, Gendreau J, Kranzler J, Rao SG. A double-blind placebo-controlled trial of milnacipran in the treatment of fibromyalgia. Human Psychopharmacology 2004;19(Suppl 1):S27-35. CENTRAL

References to studies excluded from this review

Ahmed 2016 {published data only}

Ahmed M, Aamir R, Jishi Z, Scharf MB. The effects of milnacipran on dleep fisturbance in fibromyalgia: a randomized, double-blind, placebo-controlled, two-way crossover study. Journal of Clinical Sleep Medicine 2016;12:79-86. CENTRAL

Ang 2013 {published data only}

Ang DC, Jensen MP, Steiner JL, Hilligoss J, Gracely RH, Saha C. Combining cognitive-behavioral therapy and milnacipran for fibromyalgia: a feasibility randomized-controlled trial. Clinical Journal of Pain 2013;29:747-54. CENTRAL

Branco 2011 {published data only}

Branco JC, Cherin P, Montagne A, Bouroubi A, Multinational Coordinator Study Group. Long term therapeutic response to milnacipran treatment for fibromyalgia. A European 1-year extension study following a 3-month study. Journal of Rheumatology 2011;38:1403-12. CENTRAL

Chappell 2009b {published data only}

Chappell AS, Littlejohn G, Kajdasz DK, Scheinberg M, D’Souza DN, Moldofsky H. A 1-year safety and efficacy study of duloxetine in patients with fibromyalgia. Clinical Journal of Pain 2009;25:365-75. CENTRAL

Dwight 1998 {published data only}

Dwight MM, Arnold LM, O’Brien H, Metzger R, Morris-Park E, Keck PE Jr. An open clinical trial of venlafaxine treatment of fibromyalgia. Psychosomatics 1998;39:14-7. CENTRAL

Goldenberg 2010 {published data only}

Goldenberg DL, Clauw DJ, Palmer RH, Mease P, Chen W, Gendreau RM. Durability of therapeutic response to milnacipran treatment for fibromyalgia. Results of a randomized, double-blind, monotherapy 6-month extension study. Pain Medicine 2010;11:180-94. CENTRAL

Hsiao 2007 {published data only}

Hsiao MC. Effective treatment of fibromyalgia comorbid with premenstrual dysphoric disorder with a low dose of venlafaxine. Primary Care Companion to the Journal of Clinical Psychiatry 2007;9:398. CENTRAL

Mease 2010 {published data only}

Mease PJ, Russell IJ, Kajdasz DK, Wiltse CG, Detke MJ, Wohlreich MM et al. Long-term safety, tolerability, and efficacy of duloxetine in the treatment of fibromyalgia. Seminars in Arthritis and Rheumatism 2010;39:354-64. CENTRAL

Natelson 2015 {published data only}

Natelson BH, Vu D, Mao X, Weiduschat N, Togo F, Lange G et al. Effect of milnacipran treatment on ventricular lactate in fibromyalgia: a randomized, double-blind, placebo-controlled trial. Journal of Pain 2015;16:1211-9. CENTRAL

NCT00369343 {published data only}

NCT00696787. Study Evaluating Desvenlafaxine Succinate Sustained Release (DVS SR) Versus Placebo in Peri- and Postmenopausal Women. https://clinicaltrials.gov/ct2/show/NCT00369343?term=desvenlafaxine&cond=fibromyalgia&rank=4 First posted: August 29, 2006. CENTRAL

NCT00725101 {published data only}

Allen R, Sharma U, Barlas S. Clinical experience with desvenlafaxine in treatment of patients with fibromyalgia syndrome. Clinical Pharmacology in Drug Development 2017;6:224-33. CENTRAL
NCT00725101. Fibromyalgia health outcome study on cost of treatments (REFLECTIONS). clinicaltrials.gov/ct2/results?term=NCT00725101&Search=Search First Posted: July 30, 2008. CENTRAL

NCT00793520 {published data only}

NCT00793520. Effect of milnacipran on pain processing and functional magnetic resonance imaging (fMRI) activation patterns in patients with fibromyalgia. clinicaltrials.gov/ct2/results?term=NCT00793520&Search=Search First posted: November 19,2008. CENTRAL

NCT01108731 {published data only}

NCT01108731. The effect of milnacipran in patients with fibromyalgia. clinicaltrials.gov/ct2/results?term=NCT01108731&Search=Search First posted: April 22, 2010. CENTRAL

NCT01173055 {published data only}

NCT01173055. A study to evaluate the effects of milnacipran on pain processing and functional MRI in patients with fibromyalgia. clinicaltrials.gov/ct2/results?term=NCT01173055&Search=Search First posted: July 30, 2010. CENTRAL

NCT01234675 {published data only}

NCT01234675. The effects of milnacipran on sleep disturbance in fibromyalgia. clinicaltrials.gov/ct2/results?term=NCT01234675&Search=Search First posted: November 4, 2010. CENTRAL

NCT01294059 {published data only}

NCT01294059. Effects of milnacipran on widespread mechanical and thermal hyperalgesia of fibromyalgia patients. clinicaltrials.gov/ct2/results?term=NCT01294059&Search=Search First posted: February 11, 2011. CENTRAL

NCT01331109 {published data only}

NCT01331109. Long-term safety and efficacy study of milnacipran in pediatric patients with primary fibromyalgia. clinicaltrials.gov/ct2/results?term=NCT01331109&Search=Search First posted: April 7, 2011. CENTRAL

NCT01621191 {published data only}

NCT01621191. An extension study of duloxetine in fibromyalgia (extension of F1J-JE-HMGZ, NCT01552057). clinicaltrials.gov/ct2/results?term=NCT01621191&Search=Search First posted: June 12, 2012. CENTRAL

Saxe 2012 {published data only}

Saxe PA, Arnold LM, Palmer RH, Gendreau RM, Chen W. Short-term (2-week) effects of discontinuing milnacipran in patients with fibromyalgia. Current Medical Research and Opinion 2012;28(5):815-21. CENTRAL

Sayar 2003 {published data only}

Sayar K, Aksu G, Ak I, Tosun M. Venlafaxine treatment of fibromyalgia. Annals of Pharmacotherapy 2003;37:1561-5. CENTRAL

Trugman 2014 {published data only}

Trugman JM, Palmer RH, Ma Y. Milnacipran effects on 24-hour ambulatory blood pressure and heart rate in fibromyalgia patients: a randomized, placebo-controlled, dose-escalation study. Current Medical Research & Opinion 2014;30:589-97. CENTRAL

Ziljstra 2002 {published data only}

VanderWeide, Smith SM, Trinkley KE. A systematic review of the efficacy of venlafaxine for the treatment of fibromyalgia. Journal of clinical pharmacy and therapeutics 2015;40:1-6. CENTRAL
Ziljstra TK, Barendregt PJ, Van der Laar MAF. Venlafaxine in fibromyalgia: results of a randomized placebo-controlled randomized trial. Arthritis and Rheumatism 2002;52:S105. CENTRAL

References to studies awaiting assessment

NCT00552682 {published data only}

NCT00552682. Pilot, opened, randomized clinical trial to assess the efficacy of duloxetine in the treatment of fibromialgy in patients with infection by HIV 1+. clinicaltrials.gov/ct2/results?term=NCT00552682&Search=Search First posted: November 2, 2007. CENTRAL

NCT01268631 {published data only}

NCT01268631. Mechanism-based choice of therapy: can treatments success in fibromyalgia patients be coupled to psychophysical pain modulation profile? (MTF). clinicaltrials.gov/ct2/results?term=NCT01268631&Search=Search First posted: December 31, 2010. CENTRAL

Ablin 2013

Ablin J, Fitzcharles MA, Buskila D, Shir Y, Sommer C, Häuser W. Treatment of fibromyalgia syndrome: recommendations of recent evidence-based interdisciplinary guidelines with special emphasis on complementary and alternative therapies. Evidence-based Complementary and Alternative Medicine: ECAM 2013;2013:485272.

Altman 2003

Altman DG, Bland JM. Interaction revisited: the difference between two estimates. BMJ 2003;326(7382):219.

Andersohn 2008

Andersohn F, Garbe E. Pharmacoepidemiological research with large health databases. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2008;51:1135-44.

Arnold 2010c

Arnold LM, Clauw DJ, Wohlreich MM, Wang F, Ahl J, Gaynor PJ et al. Efficacy of duloxetine in patients with fibromyalgia: pooled analysis of 4 placebo-controlled clinical trials. Primary Care Companion to the Journal of Clinical Psychiatry 2009;11(5):237-4.

Arnold 2012b

Arnold LM, Williams DA, Hudson JI, Martin SA, Clauw DJ, Crofford LJ et al. Development of responder definitions for fibromyalgia clinical trials. Arthritis and Rheumatism 2012;64:885-94.

Arnold 2013

Arnold LM, Palmer RH, Ma Y. A 3-year, open-label, flexible-dosing study of milnacipran for the treatment of fibromyalgia. Clinical Journal of Pain 2013;29:1021-8.

Arnold 2015

Arnold LM, Bateman L, Palmer RH, Lin Y. Preliminary experience using milnacipran in patients with juvenile fibromyalgia: lessons from a clinical trial program. Pediatric Rheumatology Online Journal 2015;13:27.

Ben‐Ami 2017

Ben-Ami Shor D, Weitzman D, Dahan S, Gendelman O, Bar-On Y, Amital D et al. Adherence and persistence with drug therapy among fibromyalgia patients: data from a large health maintenance organization. Journal of Rheumatology 2017;10:1499-1506.

Bernardy 2017

Bernardy K, Klose P, Welsch P, Häuser W. Cognitive behavioural therapies for fibromyalgia syndrome. European Journal of Pain 2017;22:in press. [DOI: 10.1002/14651858.CD009796.pub2]

BMJ

BMJ. Research. www.bmj.com/about-bmj/resources-authors/article-types/research.

Bradley 2009

Bradley LA. Pathophysiology of fibromyalgia. American Journal of Medicine 2009;122(12 Suppl):S22-30.

Briley 2010

Briley M. Drugs to treat fibromyalgia - the transatlantic difference. Current Opinion in Investigational Drugs 2010;11(1):16-8.

Chang 2015

Chang MH, Hsu JW, Huang KL, Su TP, Bai YM, Yang AC et al. Bidirectional association between depression and fibromyalgia syndrome: a nationwide longitudinal study. Journal of Pain 2015;16:895-902.

Choi 2010

Choi CJ, Knutsen R, Oda K, Fraser GE, Knutsen SF. The association between incident self-reported fibromyalgia and nonpsychiatric factors: 25-years follow-up of the Adventist Health Study. The Journal of Pain 2010;11:994-1003.

Clauw 1995

Clauw DJ, Katz P. The overlap between fibromyalgia and inflammatory rheumatic disease: when and why does it occur? Journal of Clinical Rheumatology 1995;1:335-42.

Clauw 2014

Clauw DJ. Fibromyalgia: a clinical review. JAMA 2014;311:1547-55.

Cohen 1988

Cohen J. Statistical power analysis for the behavioral sciences. Hillsdale: Lawrence Erlbaum Associates, 1988.

Cording 2015

Cording M, Derry S, Phillips T, Moore RA, Wiffen PJ. Milnacipran for pain in fibromyalgia in adults. Cochrane Database of Systematic Reviews 2015, Issue 10. [DOI: 10.1002/14651858.CD008244]

de Toledo 2007

de Toledo Ferraz Alves TC, Guerra de Andrade A. Hypertension induced by regular doses of milnacipran: a case report. Pharmacopsychiatry 2007;40:41-2.

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG (editors). Chapter 9: Analysing data and undertaking meta-analyses. In: 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 handbook.cochrane.org.

Department of health & Human Services 2008

Department of health & Human Services. NDA Approval. www.accessdata.fda.gov/drugsatfda_docs/appletter/2008/022148s000ltr.pdf June 13, 2008.

Department of health & Human Services 2009

Department of health & Human Services. NDA approval. https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2009/022256ltr.pdf January 14,2009.

Derry 2017

Derry S, Wiffen PJ, Häuser W, Mücke M, Tölle TR, Bell RF et al. Oral nonsteroidal anti-inflammatory drugs for fibromyalgia in adults. Cochrane Database of Systematic Reviews 2017, Issue 3. [DOI: 10.1002/14651858.CD012332.pub2]

Dworkin 2009

Dworkin RH, Turk DC, McDermott MP, Peirce-Sandner S, Burke LB, Cowan P et al. Interpreting the clinical importance of group differences in chronic pain clinical trials: IMMPACT recommendations. Pain 2009;146(3):238-44.

EMA 2008

European Medicines Agency. Refusal assessment report for Cymbalta. International non-proprietary name/common name: (duloxetine hydrochloride) Procedure No. EMEA/H/C/572/II/26. Available at www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Assessment_Report_-_Variation/human/000572/WC500076168.pdf Accessed 30 July 2009.

EMA 2010

European Medicines Agency. Refusal assessment report for milnacipran Pierre Fabre Medicament. International non-proprietary name: milnacipran. Procedure No. EMEA/H/C/001034. www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/001034/WC500089828.pdf Accessed 30 July 30 2010.

Fayers 2014

Fayers PM, Hays RD. Don’t middle your MIDs: regression to the mean shrinks estimates of minimally important differences. Quality Life Research 2014;23:1-4.

Fuller‐Thomson 2012

Fuller-Thomson E, Nimigon-Young J, Brennenstuhl S. Individuals with fibromyalgia and depression: findings from a nationally representative Canadian survey. Rheumatology International 2012;32(4):853-62.

Furukawa 2005

Furukawa TA, Cipriani A, Barbui C, Brambilla P, Watanabe N. Imputing response rates from means and standard deviations in meta-analyses. International Clinical Psychopharmacology 2005;20:49-52.

GRADEpro GDT [Computer program]

McMaster University (developed by Evidence Prime)GRADEpro GDT. Hamilton (ON): McMaster University (developed by Evidence Prime), 2015.Available at gradepro.org.

Higgins 2003

Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003;327:557-60.

Higgins 2010

Higgins A, Nash M, Lynch AM. Antidepressant-associated sexual dysfunction: impact, effects, and treatment. Drug Healthcare and Patient Safety 2010;10:141-50.

Higgins 2011

Higgins JPT, Deeks JJ, Altman DG (editors). Chapter 16: Special topics in statistics. In: 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 handbook.cochrane.org.

Häuser 2009

Häuser W, Eich W, Herrmann M, Nutzinger DO, Schiltenwolf M, Henningsen P. Fibromyalgia syndrome: classification, diagnosis, and treatment [Fibromyalgiesyndrom: klassifikation, diagnose und therapie]. Deutsches Ärzteblatt International  2009;106(23):383-91.

Häuser 2010a

Häuser W, Kosseva M, Uceyler N, Klose P, Sommer C. Emotional, physical and sexual abuse in fibromyalgia syndrome - a systematic review with meta-analysis. Arthritis Care and Research 2011;63(8):808-20.

Häuser 2010b

Häuser W, Petzke F, Sommer C. Comparative efficacy and harms of duloxetine, milnacipran, and pregabalin in fibromyalgia syndrome. Journal of Pain 2010;6:505-21.

Häuser 2010c

Häuser W, Klose P, Langhorst J, Moradi B, Steinbach M, Schiltenwolf M et al. Efficacy of different types of aerobic exercise in fibromyalgia syndrome: a systematic review and meta-analysis of randomised controlled trials. Arthritis Research Therapy 2010;12(3):R79.

Häuser 2011

Häuser W, Bartram-Wunn E, Bartram C, Tölle TR. Placebo responders in randomized controlled drug trials of fibromyalgia syndrome: systematic review and meta-analysis. Schmerz2011;25:19-31.

Häuser 2012

Häuser W, Bartram C, Bartram-Wunn E, Tölle T. Adverse events attributable to nocebo in randomized controlled drug trials in fibromyalgia syndrome and painful diabetic peripheral neuropathy: systematic review. Clinical Journal of Pain 2012;28:437-51.

Häuser 2013 a

Häuser W, Galek A, Erbslöh-Möller B, Köllner V, Kühn-Becker H, Langhorst J et al. Posttraumatic stress disorder in fibromyalgia syndrome: prevalence, temporal relationship between posttraumatic stress and fibromyalgia symptoms, and impact on clinical outcome. Pain 2013;154:1216-23.

Häuser 2013 b

Häuser W, Urrútia G, Tort S, Uçeyler N, Walitt B. Serotonin and noradrenaline reuptake inhibitors (SNRIs) for fibromyalgia syndrome. Cochrane Database of Systematic Reviews 2013, Issue 1. [DOI: 10.1002/14651858.CD010292]

Häuser 2014

Häuser W, Henningsen P. Fibromyalgia syndrome - a somaform disorder. European Journal of Pain 2014;18:1052-9.

Häuser 2015a

Häuser W, Ablin J, Fitzcharles MA, Littlejon J, Luciano JV, Usui C et al. Fibromyalgia. Nature Reviews Disease Primers 2015;1:15022.

Häuser 2015b

Häuser W, Klose P, Welsch P, Petzke F, Nothacker M, Kopp I. Methodology of the development of the updated LONTS guidelines for long-term administration of opioids in noncancer pain. Schmerz 2015;29:8-34.

Häuser 2017

Häuser W, Clauw D, Perrot S, Fitzcharles MA. Unravelling fibromyalgia – steps towards individualized management. Journal of Pain 2018;19:125-34.

International Council for Harmonisation 2016

International Council for Harmonisation. Medical Dictionary for Regulatory Activities Version 19.1. www.meddra.org/news-and-events/news/all-translations-meddra-version-191-are-now-available 20162016.

Kyle 2010

Kyle JA, Dugan BD, Testerman KK. Milnacipran for treatment of fibromyalgia. Annals of Pharmacotherapy 2010;44(9):1422-9.

Lange 2010

Lange M, Petermann F. Influence of depression on fibromyalgia: a systematic review. Schmerz 2010;24:326-33.

Lee 2012

Lee YH, Choi SJ, Ji JD, Song GG. Candidate gene studies of fibromyalgia: a systematic review and meta-analysis. Rheumatology International 2012;32:417-26.

Lee 2016

Lee YH, Song GG. Comparative efficacy and tolerability of duloxetine, pregabalin, and milnacipran for the treatment of fibromyalgia: a Bayesian network meta-analysis of randomized controlled trials. Rheumatology International 2016;36:663-72.

Legangneux 2001

Legangneux E, Mora JJ, Spreux-Varoquaux O, Thorin I, Herrou M, Alvado G et al. Cerebrospinal fluid biogenic amine metabolites, plasma-rich platelet serotonin and [3H]imipramine reuptake in the primary fibromyalgia syndrome. Rheumatology 2001;40(3):290-6.

Liu 2016

Liu Y, Qian C, Yang M. Treatment patterns associated with ACR-recommended medications in the management of fibromyalgia in the United States. Journal of Managed Care & Specialty Pharmacy 2013;22:263-71.

Lunn 2014

Lunn MP, Hughes RA, Wiffen PJ. Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia. Cochrane Database of Systematic Reviews 1, Issue 2014. [DOI: 10.1002/14651858.CD007115]

MacFarlane 2017

Macfarlane GJ, Kronisch C, Atzeni F, Häuser W, Choy EH, Amris K et al. EULAR recommendations for management of fibromyalgia. Annals of the Rheumatic Diseases 2017;76:318-328.

Mease 2009a

Mease P, Arnold LM, Choy EH, Clauw DJ, Crofford LJ, Glass JM et al (the OMERACT Fibromyalgia Working Group). Fibromyalgia syndrome module at OMERACT 9: domain construct. Journal of Rheumatology 2009;36(10):2318-29.

Mease 2013

Mease PJ, Farmer MV, Palmer RH, Gendreau RM, Wang Y. Milnacipran combined with pregabalin in fibromyalgia: a randomized, open-label study evaluating the safety and efficacy of adding milnacipran in patients with incomplete response to pregabalin. Therapeutic Advances in Musculoskeletal Diseases 2013;5:113-26.

Moore 2008

Moore RA, Barden J, Derry S, McQuay HJ. Systematic Reviews in Pain Research: Methodology Refined. 1st edition. Washington: IASP Press, 2008.

Moore 2010a

Moore RA, Eccleston C, Derry S, Wiffen P, Bell RF, Straube S et al. "Evidence" in chronic pain--establishing best practice in the reporting of systematic reviews. Pain 2010;150(3):386-9.

Moore 2010b

Moore RA, Derry S, McQuay HJ, Straube S, Aldington D, Wiffen P et al, ACTINPAIN writing group of the IASP Special Interest Group (SIG) on Systematic Reviews in Pain Relief. Clinical effectiveness: an approach to clinical trial design more relevant to clinical practice, acknowledging the importance of individual differences. Pain 2010;149(2):173-6.

Moore 2010c

Moore RA, Straube S, Paine J, Phillips CJ, Derry S, McQuay HJ. Fibromyalgia: moderate and substantial pain intensity reduction predicts improvement in other outcomes and substantial quality of life gain. Pain 2010;149:360-4.

Moore 2012

Moore RA, Straube S, Eccleston C, Derry S, Aldington D, Wiffen P et al. Estimate at your peril: imputation methods for patient withdrawal can bias efficacy outcomes in chronic pain trials using responder analyses. Pain 2012;153:265-8.

Mork 2010

Mork PJ, Vasseljen O, Nilsen TI. Association between physical exercise, body mass index, and risk of fibromyalgia: longitudinal data from the Norwegian Nord-Trøndelag Health Study. Arthritis Care & Research 2010;62:611-7.

Mork 2012

Mork PJ, Nilsen TI. Sleep problems and risk of fibromyalgia: longitudinal data on an adult female population in Norway. Arthritis & Rheumatism 2012;64:281-4.

Murakami 2017

Murakami M, Osada K, Ichibayashi H, Mizuno H, Ochiai T, Ishida M et al. An open-label, long-term, phase III extension trial of duloxetine in Japanese patients with fibromyalgia. Modern Rheumatology 2017;27:688-95.

Nishishinya 2006

Nishishinya MB, Walitt B, Urrútia G, Mease PJ, Rodríguez A, Riera Lizardo RJ et al. Anti-depressants and centrally active agents for fibromyalgia syndrome. Cochrane Database of Systematic Reviews 2006, Issue 4. [DOI: 10.1002/14651858.CD006192]

Norman 2001

Norman GR, Sridhar FG, Guyatt GH, Walter SD. Relation of distribution- and anchor-based approaches in interpretation of changes in health-related quality of life. Medical Care 2001;39(10):1039-47.

Nüesch 2013

Nüesch E, Häuser W, Bernardy K, Barth J, Jüni P. Comparative efficacy of pharmacological and non-pharmacological interventions in fibromyalgia syndrome: network meta-analysis. Annals of Rheumatic Diseases 2013;72:955-62.

Ormseth 2010

Ormseth MJ, Eyler AE, Hammonds CL, Boomershine CS. Milnacipran for the management of fibromyalgia syndrome. Journal of Pain Research 2010;3:15-24.

Petzke 2017

Petzke F, Brückle W, Eidmann U, Heldmann P, Köllner V, Kühn T et al. General treatment principles, coordination of care and patient education in fibromyalgia syndrome: updated guidelines 2017 and overview of systematic review articles. Schmerz 2017;31:246-54.

Queiroz 2013

Queiroz LP. Worldwide epidemiology of fibromyalgia. Current Pain and Headache Reports 2013;17:356.

Rahmadi 2011

Rahmadi M, Narita M, Yamashita A, Imai S, Kuzumaki N, Suzuki T. Sleep disturbance associated with an enhanced orexinergic system induced by chronic treatment with paroxetine and milnacipran. Synapse 2011;65:652-7.

RevMan 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane CollaborationReview Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Routman 2010

Routman JS, Willig JH, Westfall AO, Abroms SR, Varshney M, Adusumilli S et al. Comparative efficacy versus effectiveness of initial antiretroviral therapy in clinical trials versus routine care. Clinical Infectious Diseases 2010;15(4):574-84.

Russell 1992

Russell IJ, Vaeroy H, Javors M, Nyberg F. Cerebrospinal fluid biogenic amine metabolites in fibromyalgia/fibrositis syndrome and rheumatoid arthritis. Arthritis and Rheumatism 1992;35(5):550-6.

Schünemann 2011

Schünemann HJ, Oxman AD, Vist GE, Higgins JPT, Deeks JJ, Glasziou P et al. Chapter 12: Interpreting results and drawing conclusions. In: Higgins JPT, Green S (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Smythe 1981

Smythe HA. Fibrositis and other diffuse musculoskeletal syndromes. Textbook of Reumathology. 1st edition. Philadelphia: WB Saunders, 1981.

Taylor 2013

Taylor D, Lenox-Smith A, Bradley A. A review of the suitability of duloxetine and venlafaxine for use in patients with depression in primary care with a focus on cardiovascular safety, suicide and mortality due to antidepressant overdose. Therapeutic Advances in Psychopharmacology 2013;3:151-61.

Tort 2012

Tort S, Urrútia G, Nishishinya MB, Walitt B. Monoamine oxidase inhibitors (MAOIs) for fibromyalgia syndrome. Cochrane Database of Systematic Reviews 2012, Issue 4. [DOI: 10.1002/14651858.CD009807]

Ursini 2010

Ursini F, Pipicelli G, Grembiale RD. Efficacy and safety of duloxetine in fibromyalgia. Clinical Therapeutics 2010;161(4):391-5.

VanderWeide 2015

VanderWeide LA, Smith SM, Trinkley KE. A systematic review of the efficacy of venlafaxine for the treatment of fibromyalgia. Journal of Clinical Pharmacy and Therapeutics 2015;40:1-6.

Voican 2014

Voican CS, Corruble E, Naveau S, Perlemuter G. Antidepressant-induced liver injury: a review for clinicians. American Journal of Psychiatry 2014;171:404-15.

Walitt 2015

Walitt B, Urrútia G, Nishishinya MB, Cantrell SE, Häuser W. Selective serotonin reuptake inhibitors for fibromyalgia syndrome. Cochrane Database of Systematic Reviews 2015, Issue 6. [DOI: 10.1002/14651858.CD011735]

Wolfe 1990

Wolfe F, Smythe HA, Yunus MB, Bennett RM, Bombardier C, Goldenberg DL et al. The American College of Rheumatology 1990 criteria for the classification of fibromyalgia. Report of the Multicenter Criteria Committee. Arthritis and Rheumatism 1990;33(12):1863-4.

Wolfe 2010

Wolfe F, Clauw DJ, Fitzcharles MA, Goldenberg DL, Katz RS, Mease P et al. The American College of Rheumatology preliminary diagnostic criteria for fibromyalgia and measurement of symptom severity. Arthritis and Rheumatism 2010;62(5):600-10.

Wolfe 2011a

Wolfe F, Häuser W, Hassett AL, Katz RS, Walitt BT. The development of fibromyalgia - I: examination of rates and predictors in patients with rheumatoid arthritis (RA). Pain 2011;152(2):291-9.

Wolfe 2011b

Wolfe F, Clauw DJ, Fitzcharles MA, Goldenberg DL, Häuser W, Katz RS et al. Fibromyalgia criteria and severity scales for clinical and epidemiological studies: a modification of the ACR preliminary diagnostic criteria for fibromyalgia. Journal of Rheumatology 2011;38(6):1113-22.

Wolfe 2013a

Wolfe F, Brähler E, Hinz A, Häuser W. Fibromyalgia prevalence, somatic symptom reporting, and the dimensionality of polysymptomatic distress: results from a survey of the general population. Arthritis Care & Research 2013;64:777-85.

Wolfe 2013b

Wolfe F, Walitt BT, Katz RS, Lee YC, Michaud KD, Häuser W. Longitudinal patterns of analgesic and central acting drug use and associated effectiveness in fibromyalgia. European Journal of Pain 2013;17:581-6.

Wolfe 2016

Wolfe F, Clauw DJ, Fitzcharles MA, Goldenberg DL, Häuser W, Katz RL et al. 2016 Revisions to the 2010/2011 fibromyalgia diagnostic criteria. Seminars in Arthritis and Rheumatism 2016;46:319-29.

Yunus 1981

Yunus M, Masi AT, Calabro JJ, Miller KA, Feigenbaum SL. Primary fibromyalgia (fibrositis): clinical study of 50 patients with matched normal controls. Seminars in Arthritis and Rheumatism 1981;11(1):151-71.

Yunus 1982

Yunus M, Masi AT, Calabro JJ, Shah IK. Primary fibromyalgia. American Family Physician 1982;25(5):115-21.

Yunus 1984

Yunus MB. Primary fibromyalgia syndrome: current concepts. Comprehensive Therapy 1984;10(8):21-8.

Yunus 2008

Yunus MB. Central sensitivity syndromes: a new paradigm and group nosology for fibromyalgia and overlapping conditions, and the related issue of disease versus illness. Seminars in Arthritis and Rheumatism 2008;37:339-52.

Üceyler 2017 a

Üçeyler N, Burgmer M, Friedel E, Greiner W, Petzke F, Sarholz M et al. Etiology and pathophysiology of fibromyalgia syndrome: updated guidelines 2017, overview of systematic review articles and overview of studies on small fiber neuropathy in FMS subgroups. Schmerz 2017;31:239-45.

Üceyler 2017 b

Üçeyler N, Sommer C, Walitt B, Häuser W. Anticonvulsants for fibromyalgia. Cochrane Database of Systematic Reviews 2017, Issue 10. [DOI: 10.1002/14651858.CD010782.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Arnold 2004

Study characteristics

Methods

Study setting: multicenter study with 18 outpatient research centers in USA

Study period: July 2001‐March 2002

Study design: parallel

Trial duration: 3‐30 days' screening, 1 week single‐blind placebo run in, 12 weeks' therapy

Participants

DLX: N = 104; 88.5% female; 88.5% white; mean age 49.9 (SD 12.3) years; pain baseline (0‐10) 6.1 (SD 1.8); 35.6% current major depression

Placebo: N = 103; 89.3% female; 85.4% white; mean age 48.3 (SD 11.3) years; pain baseline (0‐10) 6.1 (SD 1.7); 40.8% current major depression

Inclusion criteria: ACR 1990 criteria; score ≥ 4 on the pain intensity item of the FIQ; age ≥ 18 years; with and without MDD

Exclusion criteria: pain from traumatic injury or structural or regional rheumatic disease; rheumatoid arthritis, inflammatory arthritis, or autoimmune disease; unstable medical or psychiatric illness; current dysthymia, which is more resistant to treatment than major depression, or primary psychiatric disorder other than MDD; substance abuse in the last year; history of psychosis; pregnancy or breast feeding; unacceptable contraception in those of childbearing potential; involvement in disability reviews that might compromise treatment response; use of an investigational drug within 30 days; prior participation in a study of DLX; severe allergic reactions to multiple medications; intolerance to 3 psychoactive drugs or 1 SSRI; and failure to respond to 2 adequate regimens of 2 different classes of antidepressants for depression or FM. Concomitant medication exclusions included use of medications or herbal agents with CNS activity (antidepressants required a 7‐day washout prior to visit 2 except for monoamine oxidase inhibitors, which required a 14‐day washout, and fluoxetine, which required a 30‐day washout); regular use of analgesics with the exception of acetaminophen up to 2 g/d and aspirin up to 325 mg/d; chronic use of sedatives, antiemetics, or antispasmodics; episodic use of anticoagulants; 3 months' stable therapy with antihypertensives, hormones, anti‐arrhythmics, antidiarrhoeals, antihistamines, cough/cold preparations (excluding dextromethorphan), or laxatives; and initiation of or change in unconventional or alternative therapies

Interventions

DLX 120 mg. Titration from 20 mg/d to 60 mg twice/day during first 2 weeks of the therapy phase, as follows: 20 mg every day for 5 days, 20 mg twice/day for at least 3 days, 40 mg twice/day for at least 2 days, and 60 mg twice a day for the remainder of the study

Placebo (N = 103)

Rescue and/or allowed medication: acetaminophen (paracetamol) up to 2 g/d and aspirin up to 325 mg/d

Outcomes

Pain: BPI average pain severity (NRS 0‐10); pain relief of ≥ 30% and ≥ 50% reported

PGIC much or very much improved: not reported; average scores of PGIC (1‐7) reported

Fatigue: FIQ single item (VAS 0‐10)

Sleep problems: BPI (NRS 0‐10): not reported

HRQoL: FIQ total score (0‐80)

AEs: physical examination, ECGs, and laboratory analysis. Further details of assessment of AEs not reported. Frequency of nausea, somnolence and insomnia insufficiently reported (not suited for meta‐analysis)

Depression: BDI ‐II total score (NRS 0‐63)

Anxiety: BAI total score (NRS 0‐63)

Disability: BPI interference from pain (NRS 0‐10)

Cognitive disturbances: not assessed

Sexual function: not assessed

Tenderness: mean tender point threshold (kg/cm²)

Notes

Conflicts of interest: Drs Crofford and Arnold have received consulting fees or honoraria in the last 2 years from Eli Lilly and Company (Dr.Crawford USD 10,000, Dr. Arnold USD 10,000). In addition to the authors employed by Eli Lilly and Company listed above, Dr. Goldstein’s wife is employed by Eli Lilly and Company.

Funding: Eli Lilly

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence using an interactive response system

Allocation concealment (selection bias)

Low risk

Central independent unit (details reported on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar, details reported on request)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; participants were adequately blinded to intervention. Blinding of outcome assessors of safety adequately described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation using LOCF for efficacy data. ITT analysis

Selective reporting (reporting bias)

High risk

Outcomes: sleep problems, 30% pain reduction and SAEs not reported

Group similarity at baseline

Low risk

No significant group differences in demographic and clinical data at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Arnold 2005

Study characteristics

Methods

Study setting: multicenter study with 21 outpatient research centers in USA

Study design: parallel

Study period: November 2002‐October 2003

Duration therapy: screening duration not reported: 12 weeks' therapy

Participants

Total sample: 100% female; 89.5% white; mean age 49.6 (SD 10.9) years; 26% current major depression

"No significant differences among treatment groups were observed in any of the patient demographics or clinical characteristics including origin, age, gender, height, weight, primary diagnoses of major depressive disorder, or secondary diagnosis of anxiety"

DLX 60 mg/d: N = 118; pain baseline (0‐10) 6.4 (SD 1.4)

DLX 120 mg/d: N = 116; pain baseline (0‐10) 6.4 (SD 1.4)

Placebo: N = 120; pain baseline (0‐10) 6.5 (SD 1.5)

Inclusion criteria: ACR 1990 criteria; score ≥ 4 on the pain intensity item of the FIQ; age ≥ 18 years; with and without MDD

Exclusion criteria: pain from traumatic injury or structural or regional rheumatic disease; rheumatoid arthritis, inflammatory arthritis, or autoimmune disease; unstable medical or psychiatric illness; current primary psychiatric diagnosis other than MDD, a primary anxiety disorder within the past year (specific phobias allowed); substance abuse within the past year; serious suicide risk; pregnancy or breast‐feeding; women who, in the opinion of the investigator, were treatment refractory or may have had an involvement in disability reviews that might compromise treatment response; severe allergic reactions to multiple medications; or prior participation in a study of DLX. Concomitant medication exclusions included use of medications or herbal agents with CNS activity; regular use of analgesics with the exception of acetaminophen up to 2 g/d and aspirin for cardiac prophylaxis up to 325 mg/d; chronic use of sedatives, antiemetics, or antispasmodics; and initiation of or change in unconventional or alternative therapies

Interventions

DLX 1‐week, double‐blind, study‐drug tapering phase at which time dosage of study drug was reduced to DLX 30 mg/d for DLX 60 mg/d‐treated participants and DLX 60 mg/d for DLX120 mg/d‐treated participants; forced titration from 60 mg/d to 120 mg/d within 3 days

Placebo

Rescue and or allowed medication: acetaminophen up to 2 g/d and aspirin up to 325 mg/d

Outcomes

Pain: (BPI average pain severity (NRS 0‐10)

PGIC much or very much improved: not reported; average scores of PGIC (1‐7) reported

Fatigue: FIQ (VAS 0‐10): not reported

Sleep problems: BPI sleep interference (NRS 0‐10)

HRQoL: FIQ total score (0‐80)

AEs: physical examination, ECGs, and laboratory analysis. Details of assessment of AEs not reported. Frequency of nausea and somnolence reported; frequency insomnia not reported

Depression: HDRS (NRS 0‐52)

Anxiety: FIQ (VAS 0‐10): not reported

Disability: BPI interference from pain (NRS 0‐10)

Sexual function: not assessed

Cognitive disturbances: not assessed

Tenderness: mean tender point threshold (kg/cm²)

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: not reported

Funding: Eli Lilly

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence using an interactive response system (details reported on request)

Allocation concealment (selection bias)

Low risk

Central independent unit (details reported on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar, details reported on request)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; participants were adequately blinded to intervention. Blinding of outcome assessors of safety adequately described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation using LOCF for efficacy data. ITT analysis

Selective reporting (reporting bias)

High risk

Outcomes of anxiety and fatigue not reported

Group similarity at baseline

Low risk

No significant group differences in demographic and clinical data at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Arnold 2010a

Study characteristics

Methods

Study setting: multicenter study with 48 outpatient research centers in USA and Puerto Rico

Study period: June 2008‐July 2009

Study design: parallel

Trial duration: 5‐30‐day screening phase, the acute phase was 12 weeks’ duration, double‐blind, and placebo‐controlled. After Week 12, participants in the placebo group were transitioned to active treatment and all participants continued for an additional 12 weeks of double‐blind treatment. An optional 2‐week drug‐tapering phase was offered at the end of the 12‐week continuation phase or for participants who discontinued early after receiving at least 2 weeks of study medication.

Participants

DLX: N = 263; 92.8% women; 77.6% white; mean age 50.7 (SD 11.3); pain baseline (0‐10) 6.5 (SD 1.5); 16.7% current major depression; 7.2% current GAD

Placebo: N = 267; 93.6% women; 77.2% white; mean age 49.6 (SD 10.8); pain baseline (0‐10) 6.5 (SD 1.6); 19.9% current major depression; 9.0% current GAD

Inclusion criteria: ACR 1990 criteria; score ≥ 4 on the pain intensity item of the FIQ; age ≥ 18 years; with and without MDD/GAD

Exclusion criteria: current or diagnosed within the past year with any primary psychiatric disorder other than MDD or GAD defined by DSM‐IV; clinically judged to be at serious risk of suicide; had any unstable medical illness likely to require intervention or hospitalization; pain symptoms unrelated to FM that could interfere with interpretation of outcome measures; regional pain syndromes; multiple surgeries or failed back syndrome; a confirmed current or previous diagnosis of rheumatoid arthritis, inflammatory arthritis, or other autoimmune disease; severe liver disease; pregnant or breast‐feeding; or history of substance abuse within the past year. Participants were also excluded if they had been treated with an adequate trial of DLX and did not respond or could not tolerate DLX; were judged by the opinion of the investigator to be treatment‐refractory in FM; or whose treatment response might be compromised by disability compensation issues.

Interventions

DLX was initiated at 30 mg and escalated to 60 mg after 1 week. At week 4 and week 8 visits, DLX dose was automatically escalated via IVRS by 30 mg daily for those participants who had < 50% reduction from baseline in their BPI 24‐h pain score and the investigator had endorsed a dose increase. If the participant could not tolerate the dose increase, it was reduced to the pre‐escalation dose via IVRS

Placebo

Rescue and/or allowed medication: acetaminophen up to 2 g/d and aspirin up to 325 mg/d

Outcomes

Pain: BPI 24‐h average pain severity (NRS 0‐10)

PGIC much or very much improved: not reported; average scores of PGIC (1‐7) reported

Fatigue: MFI general fatigue (NRS 4‐20)

Sleep problems: bothered by sleep difficulties (NRS 0‐10): data extracted from figure

HRQoL: SF‐36 physical component summary score (100‐0)

AEs: physical examination, ECGs, and laboratory analysis. Details of assessment of adverse symptoms not reported. Frequency of nausea, dizziness and insomnia reported

Depression: BDI total score (NRS 0‐63)

Anxiety: BAI total score (NRS 0‐63)

Disability: BPI pain interference pain (NRS 0‐10)

Sexual function: not assessed

Cognitive disturbances: MFI mental fatigue (NRS 4‐20)

Tenderness: not assessed

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: not declared

Funding: the study authors thank the Clinical Operations and Data Management teams of Lilly USA

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence using an interactive response system

Allocation concealment (selection bias)

Low risk

Central independent unit (details reported on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar‐details reported on request)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; patients were adequately blinded to intervention. Blinding of outcome assessors of safety adequately described.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation using LOCF for efficacy data. ITT analysis

Selective reporting (reporting bias)

Low risk

All outcomes reported

Group similarity at baseline

Low risk

No significant group differences in demographic and clinical data at baseline

Sample size bias

Low risk

> 200 participants per treatment arm

Arnold 2010b

Study characteristics

Methods

Study setting: multicenter study with 68 outpatient research centers in USA and Canada

Study period: April 2006‐June 2008

Study design: parallel

Trial duration: screening and washout (1‐4 weeks),baseline assessment (2 weeks), randomization/flexible dose escalation (4‐6 weeks), stable dose (12 weeks), and randomized discontinuation (2 weeks)

Participants

MLN: N = 516; 96.6% female; 91.9% white; mean age 49.1 (SD 10.8) years; pain baseline (0‐100) 63.1 (SD 12.5)

Placebo: N = 509; 93.7% female; 90.0% white; mean age 48.7 (SD 10.6) years; pain baseline (0‐10) 64.4 (SD 12.7)

Inclusion criteria: 1990 ACR criteria, 18‐70 years, raw score of ≥ 4 on the FIQ

Exclusion criteria: previous exposure to MLN; treatment with an investigational drug within 30 days of screening; BDI score > 25 at screening or randomization; current major depressive episode as determined by MINI; significant risk of suicide according to investigator’s judgment or results of the MINI or BDI; lifetime history of psychosis, hypomania, or mania; substance abuse; other severe psychiatric illness as determined by investigator judgment; history of behavior that would, in the investigator’s judgment, prohibit compliance for the duration of the study; active or pending disability claim, workman’s compensation claim, or litigation; pregnancy or breastfeeding; unacceptable contraception; active or unstable medical illness; prostate enlargement or other genitourinary disorder

Interventions

MLN flexible up to 100 mg/d (516 participants): MLN 12.5 mg on days 1‐3; MLN 25 mg (12.5 mg twice daily) for 4 days; MLN 50 mg (25 mg twice daily) for 7 days; MLN 75 mg (37.5 mg twice daily) for 7 days; and MLN 100 mg (50 mg twice daily) for 7 days. If side effects developed, the dose of MLN could be temporarily reduced.

Placebo

Rescue and allowed medication: tramadol or hydrocodone between randomization and week 4 (end of dose escalation). Permitted analgesic medications were acetaminophen, aspirin, and NSAIDs

Outcomes

Pain: PED 24‐h recall pain score (VAS 0‐100)

PGIC much or very much improved: reported

Fatigue: MFI total (NRS 20‐100)

Sleep problems: BPI sleep interference: not reported

HRQoL: FIQ total score

AEs: physical examination, ECGs, and laboratory analysis. Details of assessment of adverse symptoms not reported. Frequency of nausea, dizziness and insomnia reported

Depression: BDI total score (NRS 0‐63)

Anxiety: BAI total score (NRS 0‐63)

Disability: BPI pain interference pain (NRS 0‐10)

Sexual function: not assessed

Cognitive disturbances: MASQ cognitive function (NRS 38‐190)

Tenderness: not assessed

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: not declared

Funding: the study was financially supported by Forest Laboratories, Inc. Forest Laboratories, Inc. and
Cypress Bioscience, Inc. were responsible for the design and conduct of the study.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence using an interactive response system

Allocation concealment (selection bias)

Low risk

Central independent unit

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar, details reported on request)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; participants were adequately blinded to intervention. Blinding of outcome assessors of safety adequately described.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation using LOCF for efficacy data. ITT analysis

Selective reporting (reporting bias)

High risk

Outcomes of sleep and anxiety not reported

Group similarity at baseline

Low risk

No significant differences in clinical and demographic variables at baseline

Sample size bias

Low risk

> 200 participants per treatment arm

Arnold 2012a

Study characteristics

Methods

Study setting: multicenter study with 29 outpatient research centers in USA, Mexico, Israel and Argentina

Study period: September 2009‐November 2010

Study design: parallel

Trial duration: 5‐30 days' screening and wash out; 12 weeks' therapy

Participants

DLX (N = 155): 94% women, 94% white, mean age 50.9 (SD 11.9) years, pain baseline (0‐10) 6.5 (SD 1.5); 20.6% with major depressive disorder

Placebo (N = 153): 96% women, 89% white, mean age 50.7 (SD 12.5) years; pain baseline (0‐10) 6.4 (SD 1.7); 24.2% with major depressive disorder

Inclusion criteria: ACR 1990 criteria; score ≥ 4 on the pain severity item of the BPI; age ≥ 18 years; with and without MDD

Exclusion criteria: prior treatment with DLX; prior participation in a DLX study; a history of substance abuse within the past year; a primary psychiatric disorder other than MDD or GAD within the last year; a history of psychosis or bipolar disorder; clinically judged to be at risk of suicide; pregnant or breast‐feeding; pain symptoms unrelated to FM that could interfere with interpretation of outcome measures; regional pain syndromes; failed back syndrome; chronic localized pain related to any past surgery, and a confirmed current or previous diagnosis of rheumatoid arthritis; inflammatory arthritis, or infectious arthritis; or an autoimmune disease. Participants who, in the opinion of the investigator, were judged to be treatment‐refractory or whose response might be compromised by disability compensation, or had an unstable medical condition were also excluded. Concomitant medication exclusions included use of medications or herbal agents with primarily cCNS activity; regular use of analgesics other than acetaminophen up to 2 g/d and aspirin up to 325 mg for cardiac prophylaxis; topical lidocaine or capsaicin, antidepressants, anticonvulsants, barbiturates, muscle relaxants; chronic use of anti‐emetics, hypnotics and sedatives; < 3 months stable therapy of antihypertensives, anti‐arrhythmics, diuretics, and hormones; steroids other than episodic treatment of symptoms unrelated to FM; and benzodiazepine use for FM pain

Interventions

DLX 30 mg

Placebo

Rescue and/or allowed medication: some analgesics, such as non‐steroidal antiinflammatory
drugs and narcotics, were allowed episodically but only for acute injury or surgery

Outcomes

Pain: BPI) average pain severity (NRS 0‐10)

PGIC much or very much improved: not reported; average PGI assessed and reported

Fatigue: FIQ single item (VAS 0‐10): not reported

Sleep problems: not assessed

Health‐related Quality of life: FIQ total score (0‐80)

AEs: vital signs, laboratory analyses, and the C‐SSRS. Frequency of nausea, somnolence and insomnia reported

Depression: BDI ‐II total score (NRS 0‐63)

Anxiety: BAI total score (NRS 0‐63)

Disability: BPI interference from pain (NRS 0‐10)

Sexual function: not assessed

Cognitive disturbances: MFI mental fatigue (NRS 4‐20)

Tenderness: not assessed

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: BAP and SZ are full time employees and stockholders at Eli Lilly and Company. LMA has received grants from and/or is a consultant for Eli Lilly and Company, Pfizer Inc, Cypress Bioscience Inc, Forest Laboratories, Boehringer Ingelheim, Novartis, Takedo,Grunenthal and Daiichi Sankyo.

Funding: Eli Lilly

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐ generated random sequence by IVRS

Allocation concealment (selection bias)

Unclear risk

No details reported

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Capsules were identical in appearance

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Participant‐reported outcomes; participants were adequately blinded to intervention. Blinding of outcome assessors of safety adequately described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Modified BOCF for ITT‐analysis (participants who discontinued because of an AE, the baseline value was used as the endpoint, and for all other participants, the last non‐missing, post‐baseline observation before rescue therapy (if any) was used as the endpoint)

Selective reporting (reporting bias)

High risk

Study protocol available at clinicaltrials.gov; NCT00965081. All predefined outcomes except fatigue reported

Group similarity at baseline

Low risk

No significant differences in demographic and clinical variables at baseline

Sample size bias

Unclear risk

100‐199 participants per treatment arm

Bateman 2013

Study characteristics

Methods

Study setting: multicenter study with 29 outpatient research centers in USA, Mexico, Israel and Argentina

Study period: March‐December 2010

Study design: parallel

Trial duration: 2‐week, open‐label run‐in, 10 weeks' therapy

Participants

MLN (N = 79): 92% women, 94% white, mean age 48.6 (SD 10.2) years; pain baseline (0‐100) 65.4 (13.2)

Placebo (N = 21): 91% women, 95% white, mean age 48.5 (SD 11.3) years; pain baseline (0‐100) 65.2 (12.2)

Inclusion criteria: female and male outpatients, aged 18–70 years, with a diagnosis of FM who had been receiving the recommended dosage of DLX 60 mg/d (ie, the maximum dosage recommended by the US Food and Drug Administration)17 at stable doses for at least 4 weeks prior to screening. DLX prescriptions had to be for the management of FM and not for the treatment of depression or another pain syndrome. Participants with a 1‑week VAS pain recall score ≥ 40 mm to ≤ 90 mm at screening were entered into the open‐label, run‑in period of this study and continued receiving DLX 60 mg/d for an additional 2 weeks in order to confirm that they were not having an adequate response to DLX under study conditions. After this 2‐week run‐in period, participants who continued to have a VAS pain score ≥ 40 mm and who still expressed dissatisfaction with treatment were eligible for randomization. A deliberately general question was used to evaluate treatment satisfaction (ie, “Are you satisfied with DLX treatment?”) in order to allow for any potential dissatisfaction (e.g. unsatisfactory improvement in pain or non‐pain symptoms, poor tolerability, simple desire to try a new medication), as might occur in clinical practice.)

Exclusion criteria: history or current diagnosis of serious psychiatric disorder; substantial alcohol use or abuse; behavior that would, in the investigator’s judgment, prohibit participation in the study; serious suicide risk; BDI 22‐25; pregnancy or breastfeeding; unacceptable contraception in those of childbearing potential; untreated hypertension; cardiovascular disease, including myocardial infarction or stroke within the past 6 months; sitting mean systolic BP .160 mmHg or diastolic BP.100 mmHg; active or unstable medical illness; evidence of active liver disease; prostate enlargement or other genitourinary disorders; renal impairment (creatinine clearance, 30 mL/min); uncontrolled narrow‐angle glaucoma; body mass index ≥ 45 kg/m2. Excluded concomitant medications included drugs with CNS activity, such as antidepressants, anorectics, antiepileptic agents, opiates, and related analgesics (e.g. oxycodone, codeine, tramadol, narcotic patches), dopamine agonists, stimulants, and sodium oxybate

Interventions

MLN 100 mg

Placebo

Rescue and/or allowed medication: acetaminophen, aspirin, and NSAIDs. Participants requiring short‐term pain rescue medication were allowed opioid analgesics, but opioids were not permitted within days of scheduled study visits. Triptans were permitted for acute migraine treatment. Nonbenzodiazepine hypnotics were also allowed for participants requiring treatment of insomnia.

Outcomes

Pain: 1‐week recall pain intensity (VAS 0‐100)

PGIC much or very much improved: reported

Fatigue: FIQ single item (0‐10): not reported

Sleep problems: not assessed

HRQoL: FIQ total score (0‐100)

AEs: no details reported. Frequency of nausea, dizziness and insomnia reported

Depression: BDI ‐II total score; SD not reported

Anxiety: FIQ single item (0‐10) not reported

Disability: FIQ single item (VAS 0‐10)

Sexual function: Arizona Sexual Experience Scale (5‐30)

Cognitive disturbances: MASQ (38‐190)

Tenderness: not assessed

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: LB has received research support and speaker fees from Forest Laboratories, Inc. and Forest Research Institute, Inc. RHP, JMT, and YL are full‐time employees of Forest Research Institute, Inc., a wholly owned subsidiary of Forest Laboratories, Inc., and hold stock in the parent company.

Funding: Forest Research Institute

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details reported

Allocation concealment (selection bias)

Unclear risk

No details reported

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT analysis by LOCF

Selective reporting (reporting bias)

High risk

All predefined outcomes of study protocol NCT01077375 reported except fatigue and anxiety

Group similarity at baseline

Low risk

No significant differences in clinical and demographic variables

Sample size bias

High risk

< 50 participants in placebo arm, 79 in active treatment arm

Branco 2010

Study characteristics

Methods

Study setting: multicenter study with 89 outpatient research centers in 13 European countries

Study period: February 2006‐September 2007

Study design: parallel

Trial duration: 1‐ to 4‐week washout period from disallowed medications, eligible participants entered a 2‐week period in which they were trained in the use of the PED; 16 weeks' therapy; 2‐week follow‐up phase without treatment

Participants

MLN: N = 430; 95.1% female; ethnicity not reported; mean age 48.3 (SD 9.3) years; pain baseline (0‐100) 65.5 (SD 12.9)

Placebo: N = 446; 93.5% female; ethnicity not reported; mean age 49.2 (SD 10.3) years; pain baseline (0‐100) 65.0 (SD 12.7)

Inclusion criteria: 1990 ACR criteria; raw score ≥ 3 on physical function component of FIQ; baseline VAS pain intensity rating between 40‐90 (0‐100 scale)

Exclusion criteria: severe psychiatric illness including GAD or current major depressive episode (assessed by MINI, BDI‐27 score > 25), alcohol/substance abuse; significant cardiovascular, respiratory, rheumatoid, rheumatic, hepatic, renal, or other medical condition; systemic infection; epilepsy; active cancer; severe sleep apneas; unstable endocrine disease; active peptic ulcer or inflammatory bowel disease; prostatic enlargement or other genitourinary disorders (in male participants); pregnancy or breastfeeding; and history or behavior that would prohibit compliance for the duration of the study

Interventions

MLN: 25 mg once daily (evening dose, days 1 and 2); 25 mg twice daily (days 3–7); 50 mg (days 8–14); 50 mg (morning dose) and 100 mg (evening dose, days 15–21); and 100 mg (days 22–28). Participants then entered the 12‐week stable‐dose treatment period, followed by a 9‐day down‐titration phase and a 2‐week follow‐up phase without treatment.

Placebo

Rescue or allowed medication: not reported

Outcomes

Pain: PED 24‐h recall pain score (VAS 0‐100); 50% response rates not reported and not provided on request; calculated by imputation method

PGIC much or very much improved: reported

Fatigue: MFI total (NRS 20‐100)

Sleep problems: MOS‐Sleep Index II (NRS 0‐100)

HRQoL: FIQ total score (VAS 0‐100)

AEs: physical examination, ECGs, and laboratory analysis. AEs were assessed throughout the study based on spontaneous reporting by participants, investigators’ use of non‐leading questions, and clinical evaluation. Frequency of nausea, dizziness and insomnia reported

Depression: BDI total score (NRS 0‐63)

Anxiety: State‐Trait Anxiety Inventory (NRS 20‐80)

Disability: BPI pain interference pain (NRS 0‐10)

Sexual function: not assessed

Cognitive disturbances: MASQ cognitive function (NRS 38‐190)

Tenderness: not assessed

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: Dr. Branco has received grant support as an investigator and consultant for Pierre Fabre Médicament. Drs. Zachrisson and Perrot have served as speakers and consultants for Pierre Fabre Médicament. Dr. Mainguy is an employee and shareholder of Pierre Fabre Médicament.

Funding: Supported by Pierre Fabre Médicament, Boulogne, France

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence using an interactive response system (details provided on request)

Allocation concealment (selection bias)

Low risk

Central independent unit (details provided on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar‐details reported on request)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; participants were adequately blinded to intervention. Outcome assessors of safety were adequately blinded to the intervention

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation using LOCF for efficacy data. ITT analysis

Selective reporting (reporting bias)

High risk

50% pain reduction not reported and not provided on request; The FDA report on MLN stated that a female participant committed suicide and that this death was possibly related to MLN

Group similarity at baseline

Low risk

No significant differences in demographic and clinical variables at baseline

Sample size bias

Low risk

> 200 participants per treatment arm

Chappell 2009a

Study characteristics

Methods

Study setting: multicenter study with 36 outpatient research centers in Western Europe (Germany, Spain, Sweden, UK) and USA

Study period: September 2005‐December 2006

Study design: parallel

Trial duration: 1 week's screening, 27 weeks' therapy

Participants

DLX: N = 162; 92.0% female; 92.6% white; mean age 50.8 (SD 10.1) years; pain baseline (0‐10) 6.6 (SD 1.5); 22.2% current major depression

Placebo: N = 168; 94.6% female; 89.3% white; mean age 50.2 (SD 11.3) years; pain baseline (0‐10) 6.4 (SD 1.5); 22.6% current major depression

Inclusion criteria: ACR 1990 criteria; age ≥ 18 years; with and without MDD

Exclusion criteria: current or previous treatment with DLX; any current primary Axis I diagnosis other than MDD; pain symptoms related to traumatic injury, structural rheumatic disease, or regional rheumatic disease (such as osteoarthritis, bursitis, tendonitis); regional pain syndrome; multiple surgeries or failed back syndrome; confirmed current or previous diagnosis of rheumatoid arthritis, inflammatory arthritis, infectious arthritis, or an autoimmune disease; and serious medical illness

Interventions

DLX 60 mg/d or 120 mg/d: participants randomly assigned to the DLX 60 mg treatment group underwent a titration in which they received DLX 30 mg for 1 week before receiving DLX 60 mg for 12 weeks. At visit 8 (week 13) participants who did not have 50% reduction in the BPI‐Modified Short Form average pain score were blindly escalated to 120 mg. Those that could not tolerate this dose were allowed to return to the 60 mg dose. Participants were allowed to increase their dose to 120 mg at any time between visits 8 and 10 (weeks 13 and 23), based upon whether they had 50% reduction in their BPI average pain score. If at any time between visits 9 and 11 (weeks 18 and 27) participants had tolerability issues with the higher dose (120 mg), they were allowed to go back to the lower dose (60 mg).

Placebo

Rescue or allowed medication: not reported

Outcomes

Pain: BPI 24‐h average pain severity (NRS 0‐10)

PGIC much or very much improved: not reported; mean scores of PGIC (NRS 1‐7) reported

Fatigue: MFI general fatigue (NRS 4‐20)

Sleep problems: BPI sleep interference (NRS 0‐10): not reported

HRQoL: FIQ total score (VAS 0‐80)

AEs: physical examination, ECGs, and laboratory analysis. AEs were assessed throughout the study based on spontaneous reporting by participants. Frequency of nausea, somnolence and insomnia reported

Depression: BDI‐II total score (NRS 0‐63)

Anxiety: FIQ anxiety (VAS 0‐10): not reported

Disability: BPI pain interference (NRS 0‐10)

Sexual function: not assessed

Cognitive disturbances: MFI mental fatigue (NRS 4‐20)

Tenderness: mean tender point threshold (kg/cm²)

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: Drs. Chappell, Detke, and D’Souza are employees and stockholders of Eli Lilly and Company. Dr Wiltse is a former employee of Eli Lilly and Company. Dr Spaeth is a consultant to Allergan, Eli Lilly, Jazz, and Pierre Fabre Medicament, and is on the speaker bureaus of Eli Lilly and Pierre Fabre Medicament. Dr Bradley is a consultant for Eli Lilly, Pfizer, and Forest; has received grant/research support from the National Institutes of Health, the Agency for Healthcare Research and Quality, Eli Lilly, Pfizer, and the American Fibromyalgia Syndrome Association; has received honoraria from Eli Lilly, Pfizer, Forest, and the Society for Women’s Health Research; is a member of the speaker/advisory board for Pfizer; and has received royalties from UpToDate Rheumatology
Funding: Eli Lilly

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence stratified by major depression status within each study center

Allocation concealment (selection bias)

Low risk

Central independent unit (details reported on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar, details reported on request)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; participants were adequately blinded to intervention. Blinding of outcome assessors of safety adequately described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation using LOCF for efficacy data. ITT analysis

Selective reporting (reporting bias)

High risk

Outcomes of sleep and anxiety not reported

Group similarity at baseline

Low risk

No significant differences in clinical and demographic variables at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Clauw 2008

Study characteristics

Methods

Study setting: multicenter study with 86 outpatient research centers in USA

Study period: November 2004‐December 2006

Study design: parallel

Trial duration: After a 1‐ to 4‐week period for washout of prohibited medications, participants were trained in the use of the electronic study diary and entered a 2‐week baseline period, during which baseline efficacy and safety values were recorded; 15 weeks' therapy

Participants

MLN 100 mg/d: N = 399; 97.0% female; 94.0% white; mean age 49.5 (SD 13.5) years; pain baseline (0‐100) 64.6 (SD 13.5)

MLN 200 mg/d: N = 396; 97.0% female; 92.9% white; mean age 50.4 (SD 10.6) years; pain baseline (0‐100) 64.5 (SD 13.8)

Placebo: N = 401; 94.8% female; 93.5% white; mean age 50.7 (SD 10.4) years; pain baseline (0‐100) 65.7 (SD 13.3)

Inclusion criteria: 1990 ACR criteria; raw score ≥ 4 on the physical function component of the FIQ; baseline VAS pain intensity rating between ≥ 40 (0‐100 scale)

Exclusion criteria: severe psychiatric illness including GAD or current major depressive episode (assessed by MINI), BDI‐27 score > 25; alcohol/substance abuse; significant cardiovascular, respiratory, rheumatoid, rheumatic, hepatic, renal, or other medical condition; systemic infection; epilepsy; active cancer; severe sleep apneas; unstable endocrine disease; active peptic ulcer or inflammatory bowel disease; prostatic enlargement or other genitourinary disorders (in male participants); pregnancy or breastfeeding; and history or behavior that would prohibit compliance for the duration of the study

Interventions

MLN 100 mg/d or 200 mg/d: dose escalation within 3 weeks

Placebo

Rescue medication: hydrocodone up to 60 mg/d

Outcomes

Pain: PED 24‐h recall pain score (VAS 0‐100); 50% response rates not reported and not provided on request; calculated by imputation method

PGIC much or very much improved: reported (NRS 1‐7)*

Fatigue: MFI total (NRS 20‐100)

HRQoL: FIQ total score (VAS 0‐100)

AEs: physical examination, ECGs, and laboratory analysis. AEs were assessed throughout the study based on spontaneous reporting by participants and investigators’ observation. Frequency of nausea, dizziness and insomnia reported

Sleep problems: MOS‐Sleep Index II (NRS 0‐100)

Depression: BDI total score (NRS 0‐63)

Anxiety: FIQ anxiety (VAS 0‐10): not reported

Disability: MDHAQ disability subscale score

Sexual function: Arizona Sexual Function Scale (5‐50)

Cognitive disturbances: MASQ cognitive function (NRS 38‐190)

Tenderness: not assessed

Dropout due to lack of efficacy: reported

Notes

*Completer analysis

Conflicts of interest: Dr. Clauw has received grant supports by Bioscience, Inc., and serves as a consultant to Cypress Bioscience, Forest Laboratories, and Pierre Fabre Medicament,all of which are involved in the development of MLN for FM. He also acts as a consultant to Eli Lilly and Company, Pfizer Inc., Procter & Gamble, and Wyeth Pharmaceuticals. He has owned stock in Cypress Bioscience. Dr. Mease has received research grant support from Allergan, Inc.; Cypress Bioscience; Forest Laboratories; Fralex Therapeutics Inc.; Jazz Pharmaceuticals; Eli Lilly; Pfizer; and Wyeth. Drs. Palmer and Wang are employees of Forest Research Institute and own stock in Forest November 2008 Laboratories. Dr. Gendreau is an employee of Cypress Bioscience and owns stock in that company.

Funding: Forest Research Institute and Cypress Bioscience

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence using an interactive response system (details provided on request)

Allocation concealment (selection bias)

Low risk

Central independent unit (details provided on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar, details reported on request)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; patients were adequately blinded to intervention. Outcome assessors of safety were adequately blinded to the intervention

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation using baseline and LOCF for efficacy data. ITT analysis

Selective reporting (reporting bias)

High risk

Anxiety scores not reported and not provided on request

Group similarity at baseline

Low risk

No significant group differences in clinical and demographic variables

Sample size bias

Low risk

> 200 participants per treatment arm

Clauw 2013

Study characteristics

Methods

Study setting: 58 study centers, USA

Study period: November 2009‐June 2010

Study design: enriched enrolment randomized withdrawal

Trial duration: 4 weeks open‐label, 12 weeks of therapy of participants randomized to MLN or placebo, double‐blind) and 1 week of tapering (double blind)

Participants

MLN (N = 100): 96% women, 95.3% white, mean age 54.5 (SD 9.5) years; pain baseline (0‐100) 65.4 (SD 13.0)

Placebo (N = 50): 96% women, 94% white, mean age 54.0 (SD 8.5) years; pain baseline (0‐100) 65.7(SD 13.6)

Inclusion criteria: adults meeting the 1990 ACR criteria for FM who entered directly from a long‐term, open‐label, flexible‐dose, lead‐in study in which they received MLN 50 mg/d to 200 mg/d for up to 3.25 years. Prior to this lead‐in study, participants had received up to 15 months of treatment with MLN 100 mg/d or 200 mg/d during double‐blind studies, resulting in up to 4.5 years of MLN exposure prior to entering into the current discontinuation study.

Exclusion criteria: significant risk of suicide, history of serious psychiatric disorder, substantial alcohol use or abuse, pregnancy or breastfeeding, cardiovascular disease within the past 12 months, mean systolic BP > 180 mmHg or diastolic BP > 110 mmHg, uncontrolled narrow‐angle glaucoma, active liver disease, severe renal impairment and any other medical disorder that might preclude participation as judged by the principal investigator

Interventions

MLN 100 or 200 mg/d

Placebo

Rescue and/or allowed medication: monoamine oxidase inhibitors, stimulant medications, anorectic agents, daily opiates, sodium oxybate and anesthetic and/or opiate patches were prohibited. Although daily opiates were prohibited, intermittent use was allowed as needed, except during the 7 days before scheduled study visits.

Outcomes

Pain: 24‐h recall pain intensity (VAS 0‐100). Data for mean pain reduction extracted from figures. Time to loss of therapeutic response: < 30% reduction in VAS pain from pre‐MLN exposure or worsening of FM requiring alternative treatment: < 50% reduction in VAS pain from pre‐MLN exposure or worsening of FM requiring alternative treatment not assessed.

PGIC much or very much worse from randomization: reported

Fatigue: MFI Total Score (20‐100); worsening was defined as a 10‐point increase from randomization

Sleep problems: not assessed

HRQoL: FIQ‐R total score (0‐100). Data extracted from figures

AEs: AEs, vital signs and clinical laboratory tests were monitored for safety. Frequency of nausea reported, of somnolence and insomnia not reported

Depression: FIQ‐R depression (0‐10): not reported

Anxiety: FIQ‐R depression (0‐10): not reported

Disability: SF‐36 physical summary component score (50‐0); worsening was defined as a 6‐point decrease from randomization

Sexual function: not assessed

Cognitive disturbances: not assessed

Tenderness: not assessed

Dropout due to lack of efficacy: insufficiently reported (not suited for meta‐analysis)

Notes

Conflicts of interest: DJC has received grants and research support from Pfizer Inc and Forest Laboratories. He has been a consultant for and has served on advisory boards for Pfizer Inc, Eli Lilly and Co, Forest Laboratories, Inc, Cypress Bioscience, Inc (now Royalty Pharma), Pierre Fabre Pharmaceuticals, UCB and AstraZeneca. PJM has received research and grant funding as well as consultation fees from Forest Laboratories, Inc, Cypress Bioscience, Inc, Eli Lilly and Co, Pfizer Inc, Allergan, Inc, Wyeth Pharmaceuticals, Jazz Pharmaceuticals and Fralex Therapeutics. In addition to being full‐time employees of Forest Research Institute, Inc, a wholly owned subsidiary of the study sponsor (Forest Laboratories, Inc), RHP, JMT and YW hold stock in the parent company.

Funding: Pierre Fabre

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomization codes were generated and securely stored by Forest Research Institute, Inc (Jersey City, NJ, USA)

Allocation concealment (selection bias)

Low risk

IVRS and/or web response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Participant‐reported outcomes; no details reported about whether participants and outcome assessors of safety were adequately blinded to intervention.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

High risk

Protocol NCT01014585. Outcomes depression and anxiety not reported

Group similarity at baseline

Low risk

No significant group differences in clinical and demographic variables

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Leombruni 2015

Study characteristics

Methods

Study setting: single center (interdisciplinary FM outpatient department); Italy

Study period: 2011‐2012

Study design: parallel

Trial duration: no details on washout period reported; 12 weeks of therapy

Participants

DLX (N = 29 )

L‐carnitine (N = 22)

Total sample: 100% female; ethnicity not reported; mean age 54.0 (SD 8.5) years; duration FM 7.6 (SD 6.8) years. "There were no demographic or clinical differences between the two groups at baseline"

Inclusion criteria: female adults meeting the 1990 ACR criteria for FM as assessed by an experienced rheumatologist pain intensity > 3 on a VAS

Exclusion criteria: concomitant DSM‐IV TR axis I psychiatric syndrome including mood and anxiety disorders; pain due to trauma; rheumatic disease; autoimmune disease; contraindications to the use of DLX or acetylcarnitine; current antidepressant treatment

Interventions

DLX 30 or 60 mg/d

Acetyl‐carnitine: 3x500 mg/d

Rescue and/or allowed medication: no information provided

Outcomes

Pain: VAS 0‐10 (current pain); 30% and 50% and more pain relied calculated by imputation method

PGIC much or very much improved: Clinical Global Impression Improvement: Only average scores reported

Fatigue: FIQ subscale score not reported

Sleep problems: not assessed

HRQoL: FIQ total score not reported

AEs: "Side effects were assessed by the same psychiatrist". Frequency of nausea, somnolence and insomnia insufficiently reported (not suited for meta‐analysis).

Depression: HADS Depression Subscale 0‐21

Anxiety: HADS Anxiety Subscale 0‐21

Disability: SF‐36 physical summary component score (100‐0)

Sexual function: not assessed

Cognitive disturbances: not assessed

Tenderness: not assessed

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: not reported

Funding: no details provided

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details reported

Allocation concealment (selection bias)

Unclear risk

No details reported

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

High risk

Completer analysis

Selective reporting (reporting bias)

High risk

No study protocol available. FIQ scores not reported

Group similarity at baseline

Low risk

No significant differences in demographic and clinical variables at baseline

Sample size bias

High risk

< 50 participants per treatment arm

Matthey 2013

Study characteristics

Methods

Study setting: single‐center study, university hospital, Switzerland

Study period: September 2006‐September 2009

Study design: parallel

Trial duration: 1‐4 weeks' screening and washout; 7 weeks' therapy; down‐titration phase of 3‐9 days

Participants

MLN (N = 38): 100% women, ethnicity not reported, mean age 48.5 (SD 11.4) years; pain baseline (0‐100) 61.2 (SD 14.5)

Placebo (N = 39): 100% women, ethnicity not reported, mean age 50.9 (SD 11.4) years; pain baseline (0‐100) 63.5 (SD 15.1)

Inclusion criteria: Women ≥ 18 years old who met the 1990 ACR FMS criteria were included if the following criteria were met: signed informed consent, negative urine pregnancy test at screening and use of adequate contraception or absence of childbearing potential, willingness to withdraw from CNS‐active therapies, willingness to discontinue treatment with trigger point injections and anesthetics, and reported baseline weekly recall pain over 40 on a 0–100 mm VAS.

Exclusion criteria: severe psychiatric illness, current major depressive episode or screening BDI > 25, history of substance abuse, epilepsy, active cardiac disease, severe chronic obstructive pulmonary disease, active liver disease, renal impairment, documented autoimmune disease, current systemic infection, active cancer, active peptic ulcer or inflammatory bowel disease (irritable bowel syndrome excepted), unstable endocrine disorder, pregnancy or breastfeeding, concomitant use of psychotropic drugs (including antidepressants or phytotherapy), sympathicomimetics, long‐acting benzodiazepines, anticoagulants, antiepileptic drugs, centrally‐acting muscle relaxants, opioids, smoking (> 25 cigarettes a day)

Interventions

MLN with stepwise increase starting from 25 mg/d to 200 mg/d

Placebo

Rescue and/or allowed medication: no details reported

Outcomes

Pain: 1‐week recall pain intensity (VAS 0‐100). 30% and 50% and more pain reduction rates calculated by imputation method

PGIC much or very much improved: reported as odds ratio (not suited for meta‐analysis)

Fatigue: MFI Total Score (20‐100)

Sleep problems: MOS Sleep Index I (50‐0)

Quality of life: FIQ total score (0‐80)

AEs: no details reported. Frequency of nausea, somnolence and insomnia not reported. No reports on SAEs

Depression: BDI ‐II total score (0‐63)

Anxiety: State‐Trait Anxiety Inventory (20‐80)

Disability: FIQ single item (VAS 0‐10): not reported

Sexual function: not assessed

Cognitive disturbances: not assessed

Tenderness: pressure pain threshold

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: each author certifies that he or she, or a member of his or her immediate family, has
no commercial association, (i.e., consultancies, stock ownership, equity interest, patent/licensing arrangements, etc.) that might post a conflict of interest in connection with the submitted manuscript.

Funding: Pierre Fabre

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated list by sponsor

Allocation concealment (selection bias)

Low risk

Chronological order of the occurring visit 2 by sponsor

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported about whether participants and outcome assessors of safety were adequately blinded to intervention

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF method

Selective reporting (reporting bias)

High risk

All predefined outcomes of study protocol NCT00757679 except FIQ disability reported

Group similarity at baseline

Low risk

No significant group differences in clinical and demographic variables at baseline

Sample size bias

High risk

< 50 participants per treatment arm

Mease 2009b

Study characteristics

Methods

Study setting: multicenter study with 59 outpatient research centers in USA

Study period: October 2003‐July 2005

Study design: parallel

Trial duration: washout period (weeks 1‐4), 2‐week baseline period, 27 weeks' therapy (3 weeks' titration, 24 weeks' stable dose); down‐titration 3‐9 days

Participants

MLN 100 mg/d: N = 224: 95.1% women, 95.9% white, mean age 49.9 (SD 10.6) years; pain baseline (0‐100) 68.3 (SD 11.5)

MLN 200 mg/d: N = 441: 95.9% women, 93.4% white, mean age 49.2 (SD 11.0) years; pain baseline (0‐100) 69.4 (SD 11.9)

b N = 223: 95.5% women, 93.4% white, mean age 49.4 (SD 10.1) years; pain baseline (0‐100) 68.3 (SD 11.9)

Inclusion criteria: 1990 ACR criteria; baseline VAS pain intensity rating between ≥ 50 (0‐100 scale)

Exclusion criteria: severe psychiatric illness; current major depressive episode (as assessed by MINI); significant risk of suicide according to the investigator’s judgment; alcohol or other drug abuse; a history of significant cardiovascular, respiratory, endocrine, genitourinary, liver, or kidney disease; autoimmune disease; systemic infection; cancer or current chemotherapy; significant sleep apnoea; active peptic ulcer or inflammatory bowel disease

Interventions

MLN 100 mg/d or 200 mg/d

Placebo

Rescue medication: hydrocodone up to 60 mg/d

Outcomes

Pain: PED 24‐h recall pain score (VAS 0‐100); missing means and SDs provided on request

PGIC much or very much improved: Reported (NRS 1‐7)*

Fatigue: MFI total (NRS 20‐100); missing SDs reported on request

Sleep problems: MOS‐Sleep Index I (NRS 0‐100); missing SDs provided on request

HRQoL: FIQ total score (VAS 0‐100): missing SDs provided on request

AEs: physical examination, ECGs, and laboratory analysis. AEs were assessed throughout the study based on spontaneous reporting by participants and investigators’ observation. Frequency of nausea, somnolence and insomnia reported

Depression: BDI total score (NRS 0‐63): missing means and SDs provided on request

Anxiety: FIQ (VAS 0‐10): not reported

Disability: SF‐36 physical function (0‐50): missing means and SDs provided on request

Sexual function: Arizona Sexual Function Scale (5‐50): SD not reported. Data not suited for quantitative analysis.

Cognitive disturbances: MASQ cognitive function (NRS 38‐190)

Tenderness: not assessed

Dropout due to lack of efficacy: reported

Notes

*Completer analysis

Conflicts of interest: Dr. Mease has received research grant support from Pfizer Inc, Cypress Bioscience, Inc., Forest Laboratories, Inc., Eli Lilly and Company, Allergan, Wyeth Pharmaceuticals, Jazz Pharmaceuticals, and Fralex Therapeutics. Dr. Clauw has received grant support from Cypress Bioscience, Inc. and serves as a consultant to Cypress Bioscience, Inc, Forest Laboratories, Inc., Pierre Fabre Médicament, Pfizer Inc, Eli Lilly and Company, Wyeth Pharmaceuticals, and Proctor and Gamble. Dr. Mease was an investigator of this study and a consultant; Dr. Clauw was a consultant for this study. As consultants, Drs. Mease and Clauw were involved in the study design, analysis of results, and preparation of the manuscript. Drs. Gendreau, Rao, and Kranzler are employees of Cypress Bioscience, Inc. Drs. Chen and Palmer are employees of Forest Laboratories, Inc.

Funding: Forest Research Institute and Cypress Bioscience

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence using an interactive response system (details provided on request)

Allocation concealment (selection bias)

Low risk

Central independent unit (details provided on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar‐details reported on request)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; participants were adequately blinded to intervention. Blinding of outcome assessors of safety adequately described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation using LOCF for efficacy data. ITT analysis

Selective reporting (reporting bias)

High risk

Standard deviations of outcome 'sexual dysfunction' not reported and not provided on request

Group similarity at baseline

Low risk

No significant differences in demographic and clinical variables at baseline

Sample size bias

Low risk

> 200 participants per treatment arm

Murakami 2015

Study characteristics

Methods

Study setting: multicenter study with 42 outpatient research centers in Japan

Study period: March 2012‐December 2013

Study design: parallel

Trial duration: 1‐2 weeks' screening, 14 weeks' treatment, 1 week dose tapering, 1 week follow‐up observation

Participants

DLX (N = 191): 82% female; mean age 47.8 (SD 12.0) years; ethnicity not reported; 4.2% major depressive disorder; pain baseline (0‐10) 6.1 (SD 1.3)

Placebo (N = 195): 84% female; mean age 49.5 (SD 11.7) years; ethnicity not reported; 3.6% major depressive disorder; pain baseline (0‐10) 6.1 (SD 1.3)

Inclusion criteria: male and female outpatients aged 20‐75 years who met the ACR 1990 criteria of FM and had a BPI and average pain score ≥ 4 at visits 1 and 2

Exclusion criteria: past DLX treatment; serious or medically unstable disease, clinically significant abnormal laboratory values, or abnormal ECG findings; pain caused by non‐FM diseases; poorly controlled thyroid dysfunction; rheumatoid, inflammatory, or infectious arthritis; autoimmune disorders other than thyroid dysfunction; psychiatric disorders other than major depressive disorder within the past year; and suicidal tendencies as assessed using the C‐SSRS

Interventions

DLX: 20 mg for 1 week, followed by 40 mg for 1 week and then 60 mg for 12 weeks during the treatment phase

Placebo

Rescue medication: participants were prohibited from using analgesics and drugs with analgesic effects, including nonsteroidal antiinflammatory drugs, anticonvulsants, pregabalin, neurotropin, anesthetics, opioids, and adrenocorticosteroids. The use of analgesics for up to 3 consecutive days and for up to a total of 10 days was permitted only for the treatment of AEs. Co‐administration of acetaminophen at doses up to 1500 mg/d was permitted

Outcomes

Pain: BPI average pain score (NRS 0‐10)

PGIC much or very much improved: NRS 1‐7; average scores reported; data not suited for data entry

Fatigue: FIQ subscale fatigue (VAS 0‐10)

Sleep problems: BPI subscale sleep interference (NRS 0‐10)

HRQoL: FIQ total score (VAS 0‐80)

AEs: safety was assessed on the basis of the presence or absence and incidence of AEs and adverse drug reactions (ADRs) reported during the treatment phase until the end of the follow‐up observation phase. Additionally, laboratory tests (hematology, clinical chemistry, and urinalysis), ECG, body weight, and vital signs were measured. The presence or absence of suicidal tendencies was assessed using the C‐SSRS. Frequency of nausea, somnolence and insomnia reported

Depression: BDI II total score (NRS 0‐63)

Anxiety: FIQ (VAS 0‐10)

Disability: SF‐36 physical function (0‐100)

Sexual function: not assessed

Cognitive disturbances: not assessed

Tenderness: not assessed

Notes

Conflicts of interest: HM and TO are employees of Shionogi & Co. Ltd. LA is an employee of Eli Lilly Japan KK, MM, KO, and KN have provided consultancy services and MM and KO received compensation from Shionogi & Co Ltd. for their participation in this study. MM, KO, and KN did not receive any compensation for their input into this study. The authors confirm that there are no non‐financial competing interests to declare in relation to this article.

Funding: Forest Research Institute and Cypress Bioscience

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Web‐based patient registration system (ACRONET Corp., Tokyo, Japan) with a stochastic minimization procedure

Allocation concealment (selection bias)

Low risk

Blinding was maintained until the end of the study by the person responsible for the study drug assignment

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The drug allocation controller confirmed the study drugs were indiscernible in terms of appearance, packaging, and labeling, and mock titration of placebo pills was also performed to maintain blinding

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; participants were adequately blinded to intervention. Blinding of outcome assessors of safety adequately described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT by BOCF method

Selective reporting (reporting bias)

Low risk

All predefined outcomes of study protocol NCT01552057 reported

Group similarity at baseline

Low risk

No significant group differences in demographic and clinical variables at baseline

Sample size bias

Unclear risk

100‐199 participants per treatment arm

NCT00697787

Study characteristics

Methods

Study setting: multicenter study; 27 outpatient research centers, USA

Study period: 2006‐2008

Study design: parallel

Trial duration: 7‐30 days' screening, 7 days' single‐blind placebo run‐in, 8 weeks: study was stopped by the sponsor for "business reasons"

Participants

Desvenlafaxine (N =42): 100%female; mean age 47.8 (SD 10.5) years; ethnicity not reported; pain baseline scores not reported

Placebo (N = 40): 100% female; mean age 46.9 (SD 12.7) years; ethnicity not reported; pain baseline scores not reported

Pregabalin (N = 43): 100% female; mean age 46.7 (SD 11.7) years; ethnicity not reported; duration of FM and pain baseline not reported

Inclusion criteria: FM diagnosed according to 1990 ACR criteria

Exclusion criteria: unstable medical or psychological conditions that would compromise the participant's safety or put the subject at greater risk during study participation. Other painful conditions that may confound the diagnosis or assessment of FM; treatment with other drugs for FM within 14 days of study start or during the study

Interventions

Desvenlafaxine 200 mg/d

Pregabalin 450 mg/d

Placebo

Rescue medication: no details reported

Outcomes

Pain: pain score (NRS 0‐10) across the last 7 days: no 30% and 50% and more reduction rates reported and not calculable

PGIC much or very much improved: not assessed

Fatigue: not assessed

Sleep problems: not assessed

HRQoL: not assessed

AEs: no details reported. Frequency of nausea, somnolence and insomnia reported

Depression: not assessed

Anxiety: not assessed

Disability: not assessed

Sexual function: not assessed

Cognitive disturbances: not assessed

Tenderness: not assessed

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: not reported

Funding: Wyeth/Pfizer

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details reported

Allocation concealment (selection bias)

Unclear risk

No details reported

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT‐analysis by LOCF

Selective reporting (reporting bias)

Low risk

Data reported as outlined in protocol

Group similarity at baseline

High risk

No significant differences in demographic data at baseline; pain baseline not reported

Sample size bias

High risk

< 50 participants per treatment arm

Russell 2008

Study characteristics

Methods

Study setting: multicenter study with 38 outpatient research centers in USA and Puerto Rico

Study period: June 2005‐June 2006

Study design: parallel

Trial duration: 1 week screening, 26 weeks' therapy

Participants

DLX 20/60 mg/d: N = 79; 97.5% female; 83.5% white; mean age 50.9 (SD 11.4) years; pain baseline (0‐10) 6.8 (SD 1.6); 27.9% current major depression

DLX 60 mg/d: N = 150; 96.7% female; 84.7% white; mean age 51.8 (SD 10.6) years; pain baseline (0‐10) 6.5 (SD 1.4); 23.3% current major depression

DLX 120 mg/d: N = 147; 97.3% female; 82.6% white; mean age 51.0 (SD 10.8) years; pain baseline (0‐10) 6.4 (SD 1.6); 23.1% current major depression

Placebo: N = 144; 95.1% female; 82.6% white; mean age 50.3 (SD 10.9) years; pain baseline (0‐10) 6.6 (SD 1.7); 24.3% current major depression

Inclusion criteria: ACR 1990 criteria; score ≥ 4 on the pain intensity item of the FIQ; age ≥ 18 years; with and without MDD

Exclusion criteria: any current primary psychiatric diagnosis other than MDD; pain symptoms unrelated to FM that could interfere with interpretation of outcome measures; regional pain syndromes; multiple surgeries or failed back syndrome; a confirmed current or previous diagnosis of rheumatoid arthritis, inflammatory arthritis, or other autoimmune disease; unstable medical or psychiatric disorders; severe liver disease; current pregnancy or breast‐feeding; or a history of substance abuse within the past year. Participants who were judged by the investigator to be treatment‐refractory or whose response might be compromised by disability compensation issues in the opinion of the investigator were also excluded

Interventions

DLX 20/60 mg/d or 60 mg/d or 120 mg/d

Placebo

Rescue and/or allowed medication: acetaminophen up to 2 g/d and aspirin up to 325 mg/d

Outcomes

Pain: BPI average pain severity (NRS 0‐10)

PGIC much or very much improved: reported

Fatigue: MFI general fatigue (NRS 4‐20)

Sleep problems: BPI sleep interference (NRS 0‐10): not reported

HRQoL: FIQ score (VAS 0‐80)

AEs: physical examination, ECGs, and laboratory analysis. AEs were assessed throughout the study based on spontaneous reporting by participants. Frequency of nausea, somnolence and insomnia reported

Depression: BDI‐II total score (NRS 0‐63): incompletely reported

Sexual function: not assessed

Cognitive disturbances: MFI mental fatigue (NRS 4‐20)

Global perceived improvement: PGIC (NRS 1‐7)

Tenderness: mean tender point threshold (kg/cm²)

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: at doi:10.1016/j.pain. 2008.02.024. Page only accessible by fees or institutional access

Funding: Eli Lilly and Company and Boehringer Ingelheim GmbH

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomization: computer‐generated random sequence using an interactive response system

Allocation concealment (selection bias)

Low risk

Central independent unit (details reported on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar, details reported on request)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; participants were adequately blinded to intervention. Blinding of outcome assessors of safety adequately described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation using LOCF for efficacy data. ITT analysis

Selective reporting (reporting bias)

High risk

Outcomes of sleep, anxiety and depression not reported; depression scores only reported in ClinicalStudyResults.org for 20/60 mg DLX

Group similarity at baseline

Low risk

No significant differences in clinical and demographic variables at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Staud 2015

Study characteristics

Methods

Study setting: single‐center study, university hospital, USA

Study period: November 2009‐April 2014

Study design: Parallel

Trial duration: 6 weeks

Participants

MLN (N = 23): 91% women, ethnicity not reported, mean age 46.9 (SD 11.5) years; pain baseline (0‐10) 5.8 (SD 1.8)

Placebo (N = 23): 96% women, ethnicity not reported, mean age 47.5 (SD 12.0) years; pain baseline (0‐10) 5.1 (SD 1.8)

Inclusion criteria: adults > 18 years; the ability to give informed consent; fulfilment of the 1990 ACR criteria for FM, including widespread pain

Exclusion criteria: a relevant medical condition besides FM; current participation in another research protocol that could interfere with or influence the outcome measures of the present study; the inability to give informed consent; current use of analgesic drugs, anxiolytic drugs, antidepressants (all participants taking analgesic drugs or antidepressants before enrolment went through the appropriate washout phase before study entry); and previous treatment with MLN; study participants < 30 years of age and those who showed evidence for major depression

Interventions

MLN 50 mg twice daily

Placebo

Rescue and/or allowed medication: participants were not allowed to take any analgesics during the study except acetaminophen (365 mg ≤ 4 times daily)

Outcomes

Pain: daily pain intensity (VAS 0‐100). 30% and 50% and more pain reduction rates calculated by imputation method

PGIC much or very much improved: not assessed

Fatigue: 24‐ hours recall pain VAS 0‐10by electronic diary

Sleep problems: not assessed

HRQoL: not assessed

AEs: no details of dropout due to AEs and frequency of SAEs reported. Frequency of nausea, somnolence and insomnia insufficiently reported (not suited for meta‐analysis)

Depression: VAS 0‐100

Anxiety: VAS 0‐100

Disability: not assessed

Sexual function: not assessed

Cognitive disturbances: not assessed

Tenderness: quantitative sensory testing was performed. Tenderness was not considered as a clinical outcome measure for the intervention.

Dropout due to lack of efficacy: insufficiently reported (not suited for meta‐analysis)

Notes

Conflicts of interest: none of the authors has any financial or other relationships that might lead to a conflict of interest.

Funding: Pierre Fabre

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomization performed using Research Randomizer (www.randomizer.org)

Allocation concealment (selection bias)

Unclear risk

No details reported

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

High risk

Completer analysis

Selective reporting (reporting bias)

High risk

Study protocol NCT01294059 available; no details of dropouts due to AEs and frequency of SAEs reported

Group similarity at baseline

Low risk

No significant differences in clinical and demographic variables at baseline

Sample size bias

High risk

< 50 participants per treatment arm

Vitton 2004

Study characteristics

Methods

Study setting: multicenter study with 12 outpatient research centers in USA

Study period: not reported

Study design: parallel

Trial duration: 1‐4 weeks' wash out, 2 weeks' baseline and training, 12 weeks' therapy

Participants

MLN once a day (N = 46): 98% women, 82% white, mean age 47.4 (SD 11.6) years; pain baseline scores not reported; 16% current depression

MLN twice a day (N = 51): 98% women, 89% white, mean age 46.2 (SD 12.2) years; pain baseline scores not reported; 7% current depression

Placebo (N = 28): 96% women, 79% white, mean age 48.0 (SD 8.4) years; pain baseline scores not reported; 32% current depression

Inclusion criteria: 1990 ACR criteria; 18‐70 years

Exclusion criteria: severe psychiatric illness excluding depression; significant risk of suicide according to the investigator’s judgement; alcohol or other drug abuse; a history of significant cardiovascular, respiratory, endocrine, genitourinary, liver or kidney disease; autoimmune disease; systemic infection; cancer or current chemotherapy; significant sleep apnoea; life expectancy < 1 year; active peptic ulcer or inflammatory bowel disease

Interventions

MLN 200 mg/d, dose escalation within 3 weeks

Placebo

Rescue medication: hydrocodone up to 60 mg/d

Outcomes

Pain: PED 24‐h recall pain score (VAS 0‐100)

Global perceived improvement: not assessed

Fatigue: FIQ VAS 0‐10. Data provided on request. OC analysis

Sleep problems: Jenkins Sleep Survey total score (NRS). Data provided on request. OC analysis

HRQoL: FIQ total score (VAS 0‐80). Data provided on request. OC analysis

AEs: physical examination, ECGs, and laboratory analysis. AEs were assessed throughout the study based on spontaneous reporting by participants. Frequency of nausea, somnolence and insomnia not reported

Depression: FIQ VAS 0‐10. Data provided on request. OC analysis

Anxiety: FIQ VAS 0‐10. Data provided on request. OC analysis

Disability: FIQ VAS 0‐10. Data provided on request. OC analysis

Sexual function: not assessed

Cognitive disturbances: not assessed

Tenderness: not assessed

Dropout due to lack of efficacy: reported

Notes

Conflicts of interest: 3 authors were employed by Cypress Bioscience, 5 authors were consultants and 3 authors were shareholders for Cypress Bioscience

Funding: authors were employed by Cypress Bioscience

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence (details provided on request)

Allocation concealment (selection bias)

Low risk

Central independent unit (details provided on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar, details reported on request)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant‐reported outcomes; participants were adequately blinded to intervention. Blinding of outcome assessors of safety adequately described

Incomplete outcome data (attrition bias)
All outcomes

High risk

OC analysis

Selective reporting (reporting bias)

Low risk

All outcomes reported or provided on request

Group similarity at baseline

High risk

Pain baseline scores not reported; higher proportion of participants with depression in placebo group

Sample size bias

High risk

< 50 participants in two treatment arms, 51 in the third arm

ACR: American College of Rheumatology; AE: adverse event; BAI: Beck Anxiety Inventory; BDI: Beck Depression Inventory; BOCF: baseline observation carried forward (statistical method); BP: blood pressure; BPI: Brief Pain Inventory; bpm: beats per minute; C‐SSRS: Columbia‐Suicide Severity Rating Scale; CNS: central nervous system; DLX: duloxetine; DSM‐IV: Diagnostic and Statistical Manual of Mental Disorders, 4th edition; ECG: electrocardiogram; FIQ: Fibromyalgia Impact Questionnaire; FM: fibromyalgia; GAD: generalized anxiety disorder; HDRS: Hamilton Depression Rating Scale; HRQoL: health‐related quality of life; IVRS: Interactive Voice Respone System; ITT: intention‐to‐treat analysis; LOCF: last observation carried forward (statistical method); MASQ: Multiple Ability Self‐report Questionnaire; MDD: major depressive disorder; MDHAQ: Multi‐Dimensional Health Assessment Questionnaire; MFI: Multidimensional Fatigue Inventory: MINI: Mini International Neuropsychiatric Interview; MLN: milnacipran; MMRM: mixed‐effects model repeated measures; MOS: Medical Outcomes Study; NRS: Numerical rating scale; NSAID: non‐steroidal anti‐inflammatory drug; OC: observed cases; PED: patient electronic diary; PGIC: Patient Global Impression of Change Inventory; QT: The Q‐T interval represents the time for both ventricular depolarization and repolarization to occur, and therefore roughly estimates the duration of an average ventricular action potential in the ECG; SAE: serious adverse effect; SSR: Society of Skeletal Radiology; SSRI: selective serotonin reuptake inhibitors; VAS: visual analog scale

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Ahmed 2016

RCT with cross‐over design with MLN and placebo for 6 weeks each in 19 participants: < 20 participants

Ang 2013

21‐week RCT (parallel design) with MLN + CBT (N = 20), MLN + education (N = 19), and placebo + CBT (N = 19)

Branco 2011

1‐year extension study, double‐blind, randomized, not control arm, of 3 doses of MLN (468 participants)

Chappell 2009b

8‐week, open‐label period followed by a 52‐week, double‐blind, that were not placebo‐controlled, randomized to 1 or 2 doses of DLX (350 participants)

Dwight 1998

8‐week, open trial with venlafaxine in 15 participants

Goldenberg 2010

Not RCT: 6‐month extension study, double‐blind, randomized,no control arm, of 2 doses of MLN (449 participants)

Hsiao 2007

Not RCT: case report of 1 patient with fibromyalgia, comorbid with premenstrual dysphoric disorder with a low dose of venlafaxine

Mease 2010

Not RCT: 6‐month extension phases with no control arm of 2 RCTs with DLX (492 participants)

Natelson 2015

< 20 participants per treatment arm: RCT comparing 100 mg MLN with placebo for 8 weeks

NCT00369343

Data not suited for meta‐analysis: randomized, placebo‐controlled trial with 696 FM patients on 4 fixed oral doses of DVS SR (50 mg, 100 mg, 150 mg and 200 mg). Interim data analysis indicated that none of the 4 DVS SR treatment groups showed separation from placebo, and that there was a high placebo response rate. The study was discontinued because of the failure of DVS to meet the predefined efficacy criteria (pain reduction, health‐related quality of life (FIQ‐total score) and PGIC score). Details (means, SDs, absolute numbers) not reported

NCT00725101

No RCT: observational study without control group with 1700 participants

NCT00793520

< 20 participants per study arm: RCT with cross‐over design (5 weeks each period) and 1 participant comparing MLN with placebo

NCT01108731

< 20 participants per treatment arm: RCT comparing MLN with placebo in 17 participants each for 8 weeks

NCT01173055

Outcomes (change in pain threshold from baseline to week 6 of treatment, change in diffuse noxious inhibitory control, change in functional magnetic resonance imaging brain activation patterns during pressure stimulation) did not meet inclusion criteria of this review: RCT with cross‐over design with 6 weeks in each period comparing MLN and placebo in 22 participants

NCT01234675

< 10 participants per treatment arm: RCT with cross‐over design with 6 weeks for each period and 10 participants in MLN and 9 participants in placebo group

NCT01294059

Not RCT: 6‐week randomized trial without control group; number of participants not reported

NCT01331109

Not RCT: open‐label study with 57 pediatric participants over 53 weeks; no control group

NCT01621191

Not RCT: 50‐week extension study of NCT01552057 with DLX and no control arm with 149 participants

Saxe 2012

Study duration < 4 weeks: 2‐week randomized, placebo‐controlled withdrawal design. Participants who had originally received MLN 100 mg/d for 12 weeks were re‐randomized to continue MLN (n = 178) or switch directly to placebo (n = 178); participants originally receiving placebo continued with placebo (n = 359): study duration < 4 weeks

Sayar 2003

Not RCT: 12‐week, open trial with venlaxafine in 15 participants with no control arm

Trugman 2014

Only outcome criterion of efficacy was 24‐h blood pressure and did not meet the inclusion criteria for this review: RCT comparing 200 mg MLN versus placebo in 321 participants with FM (of whom 50% were classified to be hypertensive at baseline ) for 7 weeks

Ziljstra 2002

Study only published as abstract: RCT comparing 75 mg venlafaxine with placebo in 90 participants with FM for 6 weeks

CBT: cognitive behavioral therapy;DLX: duloxetine; DVS SR: desvenlafaxine sustained release; FIQ: Fibromyalgia Impact Questionnaire; FM: fibromyalgia; HRQoL: health‐related quality of life; MLN: milnacipran; PGIC: Patient Global Impression of Change; RCT: randomized controlled trial

Characteristics of studies awaiting classification [ordered by study ID]

NCT00552682

Methods

Pilot, open, randomized clinical trial

Participants

Fibromyalgia in people with HIV 1+; number not reported

Interventions

Duloxetine, dosage and comparator drug not reported

Outcomes

Not reported

Notes

Study completed; no data available in clinicaltrials.gov or Medline

NCT01268631

Methods

Not reported

Participants

Fibromyalgia; number not reported

Interventions

Duloxetine, dosage and comparator drug not reported

Outcomes

Not reported

Notes

The recruitment status of this study is unknown because the information has not been verified recently

Data and analyses

Open in table viewer
Comparison 1. SNRIs versus placebo in parallel and cross‐over design trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Self‐reported pain relief of 50% or greater Show forest plot

15

6918

Risk Difference (IV, Random, 95% CI)

0.09 [0.07, 0.11]

Analysis 1.1

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 1: Self‐reported pain relief of 50% or greater

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 1: Self‐reported pain relief of 50% or greater

1.1.1 Duloxetine

7

2582

Risk Difference (IV, Random, 95% CI)

0.10 [0.06, 0.14]

1.1.2 Milnacipran

8

4336

Risk Difference (IV, Random, 95% CI)

0.09 [0.06, 0.11]

1.2 PGIC much or very much improved Show forest plot

6

2918

Risk Difference (M‐H, Random, 95% CI)

0.19 [0.12, 0.26]

Analysis 1.2

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 2: PGIC much or very much improved

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 2: PGIC much or very much improved

1.2.1 Duloxetine

1

530

Risk Difference (M‐H, Random, 95% CI)

0.35 [0.27, 0.42]

1.2.2 Milnacipran

5

2388

Risk Difference (M‐H, Random, 95% CI)

0.15 [0.11, 0.19]

1.3 Withdrawal due to adverse events Show forest plot

15

7029

Risk Difference (IV, Random, 95% CI)

0.07 [0.04, 0.10]

Analysis 1.3

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 3: Withdrawal due to adverse events

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 3: Withdrawal due to adverse events

1.3.1 Desvenlafaxine

1

82

Risk Difference (IV, Random, 95% CI)

‐0.03 [‐0.09, 0.04]

1.3.2 Duloxetine

7

2642

Risk Difference (IV, Random, 95% CI)

0.05 [0.02, 0.07]

1.3.3 Milnacipran

7

4305

Risk Difference (IV, Random, 95% CI)

0.11 [0.07, 0.14]

1.4 Serious adverse events Show forest plot

13

6732

Risk Difference (IV, Random, 95% CI)

‐0.00 [‐0.01, 0.00]

Analysis 1.4

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 4: Serious adverse events

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 4: Serious adverse events

1.4.1 Desvenlafaxine

1

82

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.05, 0.05]

1.4.2 Duloxetine

6

2432

Risk Difference (IV, Random, 95% CI)

‐0.00 [‐0.01, 0.01]

1.4.3 Milnacipran

6

4218

Risk Difference (IV, Random, 95% CI)

‐0.00 [‐0.01, 0.01]

1.5 Self‐reported fatigue Show forest plot

12

6168

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

‐0.13 [‐0.18, ‐0.08]

Analysis 1.5

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 5: Self‐reported fatigue

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 5: Self‐reported fatigue

1.5.1 Duloxetine

5

1954

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

‐0.12 [‐0.21, ‐0.03]

1.5.2 Milnacpran

7

4214

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

‐0.14 [‐0.21, ‐0.07]

1.6 Self‐reported sleep problems Show forest plot

8

4547

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

‐0.07 [‐0.15, 0.01]

Analysis 1.6

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 6: Self‐reported sleep problems

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 6: Self‐reported sleep problems

1.6.1 Duloxetine

3

1382

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

‐0.20 [‐0.31, ‐0.10]

1.6.2 Milnacipran

5

3165

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

0.02 [‐0.05, 0.10]

1.7 Self‐reported health‐related quality of life Show forest plot

14

6861

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

‐0.20 [‐0.25, ‐0.15]

Analysis 1.7

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 7: Self‐reported health‐related quality of life

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 7: Self‐reported health‐related quality of life

1.7.1 Duloxetine

7

2604

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

‐0.22 [‐0.30, ‐0.13]

1.7.2 Milnacipran

7

4257

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

‐0.19 [‐0.25, ‐0.12]

1.8 Self‐reported pain relief of 30% or greater Show forest plot

15

6924

Risk Difference (IV, Random, 95% CI)

0.10 [0.08, 0.12]

Analysis 1.8

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 8: Self‐reported pain relief of 30% or greater

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 8: Self‐reported pain relief of 30% or greater

1.8.1 Duloxetine

7

2588

Risk Difference (IV, Random, 95% CI)

0.11 [0.07, 0.15]

1.8.2 Milnacipran

8

4336

Risk Difference (IV, Random, 95% CI)

0.10 [0.07, 0.13]

1.9 Self‐reported mean pain intensity Show forest plot

16

7014

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

‐0.22 [‐0.27, ‐0.17]

Analysis 1.9

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 9: Self‐reported mean pain intensity

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 9: Self‐reported mean pain intensity

1.9.1 Desvenlafaxine

1

82

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

0.16 [‐0.27, 0.59]

1.9.2 Duloxetine

7

2619

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

‐0.26 [‐0.35, ‐0.18]

1.9.3 Milncipran

8

4313

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

‐0.20 [‐0.26, ‐0.13]

1.10 Self‐reported depression Show forest plot

14

6478

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

‐0.16 [‐0.21, ‐0.11]

Analysis 1.10

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 10: Self‐reported depression

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 10: Self‐reported depression

1.10.1 Duloxetine

7

2264

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

‐0.25 [‐0.34, ‐0.17]

1.10.2 Milnacipran

7

4214

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

‐0.11 [‐0.17, ‐0.05]

1.11 Self‐reported anxiety Show forest plot

9

3533

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

‐0.08 [‐0.21, 0.05]

Analysis 1.11

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 11: Self‐reported anxiety

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 11: Self‐reported anxiety

1.11.1 Duloxetine

4

1403

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

‐0.07 [‐0.17, 0.04]

1.11.2 Milnacipran

5

2130

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

‐0.11 [‐0.36, 0.13]

1.12 Self‐reported disability Show forest plot

13

6789

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

‐0.21 [‐0.26, ‐0.16]

Analysis 1.12

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 12: Self‐reported disability

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 12: Self‐reported disability

1.12.1 Duloxetine

7

2602

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

‐0.29 [‐0.37, ‐0.21]

1.12.2 Milnacipran

6

4187

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

‐0.16 [‐0.22, ‐0.10]

1.13 Self‐reported cognitive disturbances Show forest plot

8

5444

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

‐0.16 [‐0.21, ‐0.10]

Analysis 1.13

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 13: Self‐reported cognitive disturbances

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 13: Self‐reported cognitive disturbances

1.13.1 Duloxetine

3

1360

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

‐0.27 [‐0.38, ‐0.16]

1.13.2 Milnacipran

5

4084

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

‐0.12 [‐0.18, ‐0.05]

1.14 Tenderness Show forest plot

5

1444

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

‐0.21 [‐0.33, ‐0.09]

Analysis 1.14

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 14: Tenderness

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 14: Tenderness

1.14.1 Duloxetine

4

1364

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

‐0.23 [‐0.35, ‐0.12]

1.14.2 Milnacipran

1

80

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

0.12 [‐0.31, 0.56]

1.15 Withdrawal due to lack of efficacy Show forest plot

14

6924

Risk Difference (IV, Random, 95% CI)

‐0.03 [‐0.04, ‐0.02]

Analysis 1.15

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 15: Withdrawal due to lack of efficacy

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 15: Withdrawal due to lack of efficacy

1.15.1 Desvenlafaxine

1

82

Risk Difference (IV, Random, 95% CI)

0.07 [‐0.02, 0.16]

1.15.2 Duloxetine

7

2642

Risk Difference (IV, Random, 95% CI)

‐0.04 [‐0.06, ‐0.02]

1.15.3 Milnacipran

6

4200

Risk Difference (IV, Random, 95% CI)

‐0.02 [‐0.04, ‐0.01]

1.16 Nausea Show forest plot

12

6606

Risk Difference (IV, Random, 95% CI)

0.16 [0.14, 0.19]

Analysis 1.16

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 16: Nausea

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 16: Nausea

1.16.1 Desvenlafaxine

1

82

Risk Difference (IV, Random, 95% CI)

0.04 [‐0.10, 0.18]

1.16.2 Duloxetine

6

2432

Risk Difference (IV, Random, 95% CI)

0.19 [0.15, 0.22]

1.16.3 Milnacipran

5

4092

Risk Difference (IV, Random, 95% CI)

0.15 [0.12, 0.18]

1.17 Somnolence Show forest plot

7

2514

Risk Difference (IV, Random, 95% CI)

0.05 [0.02, 0.08]

Analysis 1.17

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 17: Somnolence

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 17: Somnolence

1.17.1 Desvenlafaxine

1

82

Risk Difference (IV, Random, 95% CI)

‐0.05 [‐0.17, 0.06]

1.17.2 Duloxetine

6

2432

Risk Difference (IV, Random, 95% CI)

0.06 [0.03, 0.09]

1.18 Insomnia Show forest plot

9

5387

Risk Difference (M‐H, Random, 95% CI)

0.03 [0.01, 0.04]

Analysis 1.18

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 18: Insomnia

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 18: Insomnia

1.18.1 Desvenlafaxine

1

82

Risk Difference (M‐H, Random, 95% CI)

‐0.08 [‐0.18, 0.03]

1.18.2 Duloxetine

4

1684

Risk Difference (M‐H, Random, 95% CI)

0.04 [0.01, 0.07]

1.18.3 Milnacipran

4

3621

Risk Difference (M‐H, Random, 95% CI)

0.03 [0.01, 0.04]

Study flow diagram

Figuras y tablas -
Figure 1

Study flow diagram

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

Figuras y tablas -
Figure 2

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

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

Figuras y tablas -
Figure 3

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

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 1: Self‐reported pain relief of 50% or greater

Figuras y tablas -
Analysis 1.1

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 1: Self‐reported pain relief of 50% or greater

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 2: PGIC much or very much improved

Figuras y tablas -
Analysis 1.2

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 2: PGIC much or very much improved

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 3: Withdrawal due to adverse events

Figuras y tablas -
Analysis 1.3

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 3: Withdrawal due to adverse events

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 4: Serious adverse events

Figuras y tablas -
Analysis 1.4

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 4: Serious adverse events

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 5: Self‐reported fatigue

Figuras y tablas -
Analysis 1.5

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 5: Self‐reported fatigue

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 6: Self‐reported sleep problems

Figuras y tablas -
Analysis 1.6

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 6: Self‐reported sleep problems

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 7: Self‐reported health‐related quality of life

Figuras y tablas -
Analysis 1.7

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 7: Self‐reported health‐related quality of life

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 8: Self‐reported pain relief of 30% or greater

Figuras y tablas -
Analysis 1.8

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 8: Self‐reported pain relief of 30% or greater

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 9: Self‐reported mean pain intensity

Figuras y tablas -
Analysis 1.9

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 9: Self‐reported mean pain intensity

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 10: Self‐reported depression

Figuras y tablas -
Analysis 1.10

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 10: Self‐reported depression

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 11: Self‐reported anxiety

Figuras y tablas -
Analysis 1.11

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 11: Self‐reported anxiety

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 12: Self‐reported disability

Figuras y tablas -
Analysis 1.12

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 12: Self‐reported disability

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 13: Self‐reported cognitive disturbances

Figuras y tablas -
Analysis 1.13

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 13: Self‐reported cognitive disturbances

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 14: Tenderness

Figuras y tablas -
Analysis 1.14

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 14: Tenderness

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 15: Withdrawal due to lack of efficacy

Figuras y tablas -
Analysis 1.15

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 15: Withdrawal due to lack of efficacy

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 16: Nausea

Figuras y tablas -
Analysis 1.16

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 16: Nausea

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 17: Somnolence

Figuras y tablas -
Analysis 1.17

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 17: Somnolence

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 18: Insomnia

Figuras y tablas -
Analysis 1.18

Comparison 1: SNRIs versus placebo in parallel and cross‐over design trials, Outcome 18: Insomnia

Summary of findings 1. Serotonin noradrenaline reuptake inhibitors compared with placebo for fibromyalgia ‐ studies with parallel design

Serotonin noradrenaline reuptake inhibitors compared with placebo for fibromyalgia ‐ studies with parallel design

Patient or population: people with fibromyalgia

Settings: study centers in North, Central and South America, Asia and Europe

Intervention: serotonin noradrenaline reuptake inhibitors (duloxetine, milnacipran)

Comparison: placebo

Outcomes

Probable outcome with intervention

(95% CI)

Probable outcome with placebo

Relative effect

SMD or risk difference
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Self‐reported pain relief of 50% or greater

309 per 1000

(282 to 344)

210 per 1000

RD 0.09 (0.07 to 0.11)

6918 (15 studies)

⊕⊕⊝⊝
low1,2

NNTB 11 (95% CI 9 to 14)

Patient Global Impression to be much or very much improved (PGIC)

519 per 1000

(459 to 573)

293 per 1000

RD 0.19 (0.12 to 0.26)

2918 (6 studies)

⊕⊕⊝⊝
low1,2

NNTB 5 (95% CI 4 to 8)

Self‐reported fatigue (20‐100 scale)

Higher scores indicate higher fatigue problem levels

Mean fatigue
score was 2.6 points
lower (1.0 to
5.0 points lower) based on a 20‐100 scale

Baseline mean score 69.4 (SD 12.3)3

SMD ‐0.13 (‐0.18 to ‐0.08)

6168 (12 studies)

⊕⊕⊝⊝
low1,2

NNTB 18 (95% CI 12 to 29)

Self‐reported sleep problems

(0‐100 scale)

Higher scores indicate higher sleep problem levels

Mean sleep problems
score was 1.2 points
lower (0.2 higher to
5.5 points lower) based on a 0‐100 scale

Baseline mean score 68.0 (23.8)4

SMD ‐0.07 (‐0.15 to 0.01)

4547 (8 studies)

⊕⊕⊝⊝
low1,2

NNTB not calculated due to lack of statistically significant difference

Self‐reported health‐related quality of life (0‐100 scale)

Higher scores indicate higher burden of disease (lower quality of life)

Mean health‐related quality of life problems score was 3.9 points lower (2.3 to
5.3 points lower) based on a
0‐100 scale

Baseline mean score 57.9 (SD
14.1)5

SMD ‐0.20 (‐0.25 to ‐0.15)

6861 (14 studies)

⊕⊕⊝⊝
low1,2

NNTB 11 (95% CI 8 to 14)

Tolerability (withdrawal due to adverse events)

191 per 1000

(172 to 210)

102 per 1000

RD 0.07 (0.04 to 0.10)

7029 (15 studies)

⊕⊕⊝⊝
low1,2

NNTH 14 (95% CI 10 to 25)

Safety (serious adverse events)

18 per 1000

(16 to 20)

21 per 1000

RD ‐0.00 (‐0.01 to 0.00)

6732 (13 studies)

⊕⊝⊝⊝
Verylow1,2,6

NNTH not calculated due to lack of statistically significant difference

*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; FIQ: Fibromyalgia Impact Questionnaire; MFI: Multidimensional Fatigue Inventory; MOS‐Sleep problem index: Medical Outcome Study ‐ sleep problem index; NNTB: number needed to treat for an additional beneficial outcome; NNTH: number needed to treat for an additional harm; NRS: numerical rating scale; RD: risk difference; SMD: standardized mean difference; VAS: visual analog scale

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

1Downgraded once: indirectness: participants with major medical diseases and mental disorders except major depression excluded in > 50% of studies
2Downgraded once: publication bias
3Clauw 2008: N = 401 participants; MFI NRS 20‐100 scale
4Mease 2009b: N = 223 participants; MOS Sleep problem index NRS 0‐100 scale
5Arnold 2010b; N = 509 participants; FIQ VAS 0‐80 scale
6Downgraded once: imprecision due to low event rate

Figuras y tablas -
Summary of findings 1. Serotonin noradrenaline reuptake inhibitors compared with placebo for fibromyalgia ‐ studies with parallel design
Table 1. Subgroup analysis. Efficacy and safety of SNRIs in studies with North American and European participants

Outcome

Number

of

participants (studies)

Effect size

RD (95% CI)

Test for

overall

effect

P value

Heterogeneity

(%)

 

Test of interaction:

effect estimate and P value

Self‐reported pain relief 50% or greater

 

 

 

 

 Z = 0.78; P = 0.43

Only North American participants

3935 (8)

0.10 (0.08 to 0.13)

< 0.001

0

Only European participants

960 (2)

0.06 (0.01 to 0.12)

0.02

0

 

Withdrawal due to adverse events

 

 

 

 

 Z = 1.14; P = 0.25

Only North American participants

3935 (8)

0.08 (0.04 to 0.13)

< 0.0002

71

Only European participants

960 (2)

0.12 (0.08 to 0.17)

< 0.0001

0

 

CI: confidence interval; RD: risk difference; SNRIs: serotonin and noradrenaline reuptake inhibitors

Figuras y tablas -
Table 1. Subgroup analysis. Efficacy and safety of SNRIs in studies with North American and European participants
Comparison 1. SNRIs versus placebo in parallel and cross‐over design trials

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Self‐reported pain relief of 50% or greater Show forest plot

15

6918

Risk Difference (IV, Random, 95% CI)

0.09 [0.07, 0.11]

1.1.1 Duloxetine

7

2582

Risk Difference (IV, Random, 95% CI)

0.10 [0.06, 0.14]

1.1.2 Milnacipran

8

4336

Risk Difference (IV, Random, 95% CI)

0.09 [0.06, 0.11]

1.2 PGIC much or very much improved Show forest plot

6

2918

Risk Difference (M‐H, Random, 95% CI)

0.19 [0.12, 0.26]

1.2.1 Duloxetine

1

530

Risk Difference (M‐H, Random, 95% CI)

0.35 [0.27, 0.42]

1.2.2 Milnacipran

5

2388

Risk Difference (M‐H, Random, 95% CI)

0.15 [0.11, 0.19]

1.3 Withdrawal due to adverse events Show forest plot

15

7029

Risk Difference (IV, Random, 95% CI)

0.07 [0.04, 0.10]

1.3.1 Desvenlafaxine

1

82

Risk Difference (IV, Random, 95% CI)

‐0.03 [‐0.09, 0.04]

1.3.2 Duloxetine

7

2642

Risk Difference (IV, Random, 95% CI)

0.05 [0.02, 0.07]

1.3.3 Milnacipran

7

4305

Risk Difference (IV, Random, 95% CI)

0.11 [0.07, 0.14]

1.4 Serious adverse events Show forest plot

13

6732

Risk Difference (IV, Random, 95% CI)

‐0.00 [‐0.01, 0.00]

1.4.1 Desvenlafaxine

1

82

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.05, 0.05]

1.4.2 Duloxetine

6

2432

Risk Difference (IV, Random, 95% CI)

‐0.00 [‐0.01, 0.01]

1.4.3 Milnacipran

6

4218

Risk Difference (IV, Random, 95% CI)

‐0.00 [‐0.01, 0.01]

1.5 Self‐reported fatigue Show forest plot

12

6168

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

‐0.13 [‐0.18, ‐0.08]

1.5.1 Duloxetine

5

1954

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

‐0.12 [‐0.21, ‐0.03]

1.5.2 Milnacpran

7

4214

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

‐0.14 [‐0.21, ‐0.07]

1.6 Self‐reported sleep problems Show forest plot

8

4547

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

‐0.07 [‐0.15, 0.01]

1.6.1 Duloxetine

3

1382

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

‐0.20 [‐0.31, ‐0.10]

1.6.2 Milnacipran

5

3165

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

0.02 [‐0.05, 0.10]

1.7 Self‐reported health‐related quality of life Show forest plot

14

6861

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

‐0.20 [‐0.25, ‐0.15]

1.7.1 Duloxetine

7

2604

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

‐0.22 [‐0.30, ‐0.13]

1.7.2 Milnacipran

7

4257

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

‐0.19 [‐0.25, ‐0.12]

1.8 Self‐reported pain relief of 30% or greater Show forest plot

15

6924

Risk Difference (IV, Random, 95% CI)

0.10 [0.08, 0.12]

1.8.1 Duloxetine

7

2588

Risk Difference (IV, Random, 95% CI)

0.11 [0.07, 0.15]

1.8.2 Milnacipran

8

4336

Risk Difference (IV, Random, 95% CI)

0.10 [0.07, 0.13]

1.9 Self‐reported mean pain intensity Show forest plot

16

7014

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

‐0.22 [‐0.27, ‐0.17]

1.9.1 Desvenlafaxine

1

82

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

0.16 [‐0.27, 0.59]

1.9.2 Duloxetine

7

2619

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

‐0.26 [‐0.35, ‐0.18]

1.9.3 Milncipran

8

4313

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

‐0.20 [‐0.26, ‐0.13]

1.10 Self‐reported depression Show forest plot

14

6478

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

‐0.16 [‐0.21, ‐0.11]

1.10.1 Duloxetine

7

2264

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

‐0.25 [‐0.34, ‐0.17]

1.10.2 Milnacipran

7

4214

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

‐0.11 [‐0.17, ‐0.05]

1.11 Self‐reported anxiety Show forest plot

9

3533

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

‐0.08 [‐0.21, 0.05]

1.11.1 Duloxetine

4

1403

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

‐0.07 [‐0.17, 0.04]

1.11.2 Milnacipran

5

2130

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

‐0.11 [‐0.36, 0.13]

1.12 Self‐reported disability Show forest plot

13

6789

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

‐0.21 [‐0.26, ‐0.16]

1.12.1 Duloxetine

7

2602

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

‐0.29 [‐0.37, ‐0.21]

1.12.2 Milnacipran

6

4187

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

‐0.16 [‐0.22, ‐0.10]

1.13 Self‐reported cognitive disturbances Show forest plot

8

5444

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

‐0.16 [‐0.21, ‐0.10]

1.13.1 Duloxetine

3

1360

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

‐0.27 [‐0.38, ‐0.16]

1.13.2 Milnacipran

5

4084

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

‐0.12 [‐0.18, ‐0.05]

1.14 Tenderness Show forest plot

5

1444

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

‐0.21 [‐0.33, ‐0.09]

1.14.1 Duloxetine

4

1364

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

‐0.23 [‐0.35, ‐0.12]

1.14.2 Milnacipran

1

80

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

0.12 [‐0.31, 0.56]

1.15 Withdrawal due to lack of efficacy Show forest plot

14

6924

Risk Difference (IV, Random, 95% CI)

‐0.03 [‐0.04, ‐0.02]

1.15.1 Desvenlafaxine

1

82

Risk Difference (IV, Random, 95% CI)

0.07 [‐0.02, 0.16]

1.15.2 Duloxetine

7

2642

Risk Difference (IV, Random, 95% CI)

‐0.04 [‐0.06, ‐0.02]

1.15.3 Milnacipran

6

4200

Risk Difference (IV, Random, 95% CI)

‐0.02 [‐0.04, ‐0.01]

1.16 Nausea Show forest plot

12

6606

Risk Difference (IV, Random, 95% CI)

0.16 [0.14, 0.19]

1.16.1 Desvenlafaxine

1

82

Risk Difference (IV, Random, 95% CI)

0.04 [‐0.10, 0.18]

1.16.2 Duloxetine

6

2432

Risk Difference (IV, Random, 95% CI)

0.19 [0.15, 0.22]

1.16.3 Milnacipran

5

4092

Risk Difference (IV, Random, 95% CI)

0.15 [0.12, 0.18]

1.17 Somnolence Show forest plot

7

2514

Risk Difference (IV, Random, 95% CI)

0.05 [0.02, 0.08]

1.17.1 Desvenlafaxine

1

82

Risk Difference (IV, Random, 95% CI)

‐0.05 [‐0.17, 0.06]

1.17.2 Duloxetine

6

2432

Risk Difference (IV, Random, 95% CI)

0.06 [0.03, 0.09]

1.18 Insomnia Show forest plot

9

5387

Risk Difference (M‐H, Random, 95% CI)

0.03 [0.01, 0.04]

1.18.1 Desvenlafaxine

1

82

Risk Difference (M‐H, Random, 95% CI)

‐0.08 [‐0.18, 0.03]

1.18.2 Duloxetine

4

1684

Risk Difference (M‐H, Random, 95% CI)

0.04 [0.01, 0.07]

1.18.3 Milnacipran

4

3621

Risk Difference (M‐H, Random, 95% CI)

0.03 [0.01, 0.04]

Figuras y tablas -
Comparison 1. SNRIs versus placebo in parallel and cross‐over design trials