Scolaris Content Display Scolaris Content Display

Anticonvulsivos para fibromialgia

This is not the most recent version

Collapse all Expand all

References

References to studies included in this review

Arnold 2007 {published data only}

Arnold LM, Goldenberg DL, Stanford SB, Lalonde JK, Sandhu HS, Keck PE, et al. Gabapentin in the treatment of fibromyalgia: a randomized, double‐blind, placebo‐controlled, multicenter trial. Arthritis and Rheumatism 2007;56(4):1336‐44.

Arnold 2008 {published and unpublished data}

Arnold LM, Russell IJ, Diri EW, Duan WR, Young JP, Sharma U, et al. A 14‐week, randomized, double‐blinded, placebo‐controlled monotherapy trial of pregabalin in patients with fibromyalgia. Pain 2008;9(9):792‐805.

Crofford 2005 {published and unpublished data}

Crofford LJ, Rowbotham MC, Mease PJ, Russell IJ, Dworkin RH, Corbin AE, et al. Pregabalin 1008‐105 Study Group. Pregabalin for the treatment of fibromyalgia syndrome: results of a randomized, double‐blind, placebo‐controlled trial. Arthritis and Rheumatism 2005;52(4):264‐73.

Mease 2008 {published and unpublished data}

Mease PJ, Russell IJ, Arnold LM, Florian H, Young JP, Martin SA, et al. A randomized, double‐blind, placebo‐controlled, phase III trial of pregabalin in the treatment of patients with fibromyalgia. Journal of Rheumatology 2008;35(3):502‐14.

Ohta 2012 {published and unpublished data}

Ohta H, Oka H, Usui C, Ohkura M, Suzuki M, Nishioka K. A randomized, double‐blind, multicenter, placebo‐controlled phase III trial to evaluate the efficacy and safety of pregabalin in Japanese patients with fibromyalgia. Arthritis Research and Therapy 2012;14:R217.

Pauer 2011 {published and unpublished data}

Pauer L, Winkelmann A, Arsenault P, Jespersen A, Whelan L, Atkinson G, et al. A0081100 Investigators. An international, randomized, double‐blind, placebo‐controlled, phase III trial of pregabalin monotherapy in treatment of patients with fibromyalgia. Journal of Rheumatology 2011;38(12):2643‐52.

Rowbotham 2012 {published and unpublished data}

Rowbotham M. Levetiracetam for treatment of pain associated with fibromyalgia. First received on 15 November 2005; last updated on 12 April 2011. www.clinicaltrials.gov/ct2/show/NCT00254657 (accessed 4 October 2013).

UCB 2006 {published data only}

UCB. Assessing efficacy and safety of lacosamide compared to placebo in reducing signs and symptoms of fibromyalgia syndrome. First received on 9 November 2006; last updated 30 August 2011. clinicaltrials.gov/ct2/show/NCT00401830 (accessed 4 October 2013).

References to studies excluded from this review

Crofford 2008 {published and unpublished data}

Crofford LJ, Mease PJ, Simpson SL, Young JP, Martin SA, Haig GM, et al. Fibromyalgia Relapse Evaluation and Efficacy for Durability Of Meaningful relief (FREEDOM): a 6‐month, double‐blind, placebo‐controlled trial with pregabalin. Pain 2008;136(3):419‐31.

Roth 2012 {published data only}

Roth T, Lankford DA, Bhadra P, Whalen E, Resnick EM. Effect of pregabalin on sleep in patients with fibromyalgia and sleep maintenance disturbance: a randomized, placebo‐controlled, 2‐way crossover polysomnography study. Arthritis Care and Research (Hoboken) 2012;64:597‐606.

Additional references

Arnold 2010a

Arnold LM, Leon T, Whalen E, Barrett J. Relationships among pain and depressive and anxiety symptoms in clinical trials of pregabalin in fibromyalgia. Psychosomatics 2010;51:489‐57.

Arnold 2012

Arnold LM, Emir B, Murphy TK, Zeiher BG, Pauer L, Scott G, et al. Safety profile and tolerability of up to 1 year of pregabalin treatment in 3 open‐label extension studies in patients with fibromyalgia. Clinical Therapeutics 2012;34:1092‐102.

Attal 2010

Attal N, Cruccu G, Baron R, Haanpää M, Hansson P, Jensen TS, et al. European Federation of Neurological Societies. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. European Journal of Neurology 2010;17:1117‐e88.

Begg 1994

Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias. Biometrics 1994;50(4):1088‐101.

Bennett 2007

Bennett RM, Jones J, Turk DC, Russell IJ, Matallana L. An internet survey of 2,596 people with fibromyalgia. BMC Musculoskeletal Disorders 2007;8:27.

Bernardy 2010

Bernardy K, Füber N, Köllner V, Häuser W. Efficacy of cognitive‐behavioral therapies in fibromyalgia syndrome ‐ a systematic review and metaanalysis of randomized controlled trials. Journal of Rheumatology 2010;37(10):1991‐2005.

Bernardy 2013

Bernardy K, Klose P, Busch AJ, Choy EHS, Häuser W. Cognitive behavioural therapies for fibromyalgia. Cochrane Database of Systematic Reviews 2013, Issue 9. [DOI: 10.1002/14651858.CD009796]

Beydoun 2009

Beydoun A, D'Souza J, Hebert D, Doty P. Lacosamide: pharmacology, mechanisms of action and pooled efficacy and safety data in partial‐onset seizures. Expert Review of Neurotherapeutics 2009;9(1):33‐42.

Birse 2012

Birse F, Derry S, Moore RA. Phenytoin for neuropathic pain and fibromyalgia in adults. Cochrane Database of Systematic Reviews 2012, Issue 5. [DOI: 10.1002/14651858.CD009485.pub2]

Bockbrader 2010

Bockbrader HN, Wesche D, Miller R, Chapel S, Janiczek N, Burger P. A comparison of the pharmacokinetics and pharmacodynamics of pregabalin and gabapentin. Clinical Pharmacokinetics 2010;49:661‐9.

Boomershine 2010

Boomershine CS. Pregabalin for the management of fibromyalgia syndrome. Journal of Pain Research 2010;22:81‐8.

Bradley 2009

Bradley LA. Pathophysiology of fibromyalgia. American Journal of Medicine 2009;122:S22‐S30.

Branco 2010

Branco JC, Bannwarth B, Failde I, Abello Carbonell J, Blotman F, Spaeth M, et al. Prevalence of fibromyalgia: a survey in five European countries. Seminars in Arthritis and Rheumatism 2010;39(6):448‐53.

Briley 2010

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

Buskila 2009

Buskila D. Developments in the scientific and clinical understanding of fibromyalgia. Arthritis Research Therapy 2009;11:242.

Centers for Disease Control and Prevention 2011

Centers for Disease Control and Prevention, 2012. Fibromyalgia. www.cdc.gov/arthritis/basics/fibromyalgia.htm (accessed 4 October 2013).

Cohen 1988

Cohen J. Statistical Power Analysis for the Behavioral Sciences. Hillsdale: Lawrence Erlbaum Associates, 1988.

Corrigan 2012

Corrigan R, Derry S, Wiffen PJ, Moore RA. Clonazepam for neuropathic pain and fibromyalgia in adults. Cochrane Database of Systematic Reviews 2012, Issue 5. [DOI: 10.1002/14651858.CD009486.pub2]

Deare 2013

Deare JC, Zheng Z, Xue CCL, Liu JP, Shang J, Scott SW, et al. Acupuncture for treating fibromyalgia. Cochrane Database of Systematic Reviews 2013, Issue 5. [DOI: 10.1002/14651858.CD007070.pub2]

Derry 2012

Derry S, Gill D, Phillips T, Moore RA. Milnacipran for neuropathic pain and fibromyalgia in adults. Cochrane Database of Systematic Reviews 2012, Issue 3. [DOI: 10.1002/14651858.CD008244.pub2]

Dooley 2007

Dooley DJ, Taylor CP, Donevan S, Feltner D. Ca2+ channel alpha2delta ligands: novel modulators of neurotransmission. Trends in Pharmacological Sciences 2007;28(2):151.

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.

Eccleston 2012

Eccleston C, Palermo TM, Williams ACDC, Lewandowski A, Morley S, Fisher E, et al. Psychological therapies for the management of chronic and recurrent pain in children and adolescents. Cochrane Database of Systematic Reviews 2012, Issue 12. [DOI: 10.1002/14651858.CD003968]

Egger 1997

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

Eich 2012a

Eich W, Häuser W, Arnold B, Bernardy K, Brückle W, Eidmann U, et al. Fibromyalgia syndrome: general principles and coordination of clinical care and patient education. Schmerz 2012;26:268‐75.

Eich 2012b

Eich W, Häuser W, Arnold B, Jäckel W, Offenbächer M, Petzke F, et al. Fibromyalgia syndrome: definition, classification, clinical diagnosis and prognosis. Schmerz 2012;26:247‐58.

European Medicines Agency 2009

European Medicines Agency. Refusal assessment report for Lyrica, 2009. www.ema.europa.eu/docs/en_GB/document_library/EPAR_‐_Assessment_Report_‐_Variation/human/000546/WC500076177.pdf (accessed 5 October 2013).

FDA 2007

FDA. FDA approves first drug for treating fibromyalgia, 2007. www.fda.gov/newsevents/newsroom/pressAnnouncements/2007/ucm108936.htm (accessed 4 October 2013).

Fitzcharles 2013

Fitzcharles MA, Ste‐Marie PA, Goldenberg DL, Pereira JX, Abbey S, Choinière M, et al. Canadian Pain Society and Canadian Rheumatology Association recommendations for rational care of persons with fibromyalgia. A summary report. Journal of Rheumatology 2013;40:1388‐93.

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.

Furukawa 2006

Furukawa TA, Barbui C, Cipriani A, Brambilla P, Watanabe N. Imputing missing standard deviations in meta‐analyses can provide accurate results. Journal of Clinical Epidemiology 2006;59:7‐10.

Gahr 2013

Gahr M, Freudenmann RW, Hiemke C, Kölle MA, Schönfeldt‐Lecuona C. Pregabalin abuse and dependence in Germany: results from a database query. European Journal of Clinical Pharmacology 2013;69(6):1335‐42.

Gracely 2011

Gracely RH, Ambrose KR. Neuroimaging of fibromyalgia. Best Practice and Research Clinical Rheumatology 2011;25(2):271‐84.

Guyatt 2011

Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, et al. GRADE guidelines: 1. Introduction‐GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 2011;64(4):383‐94.

Hearn 2012

Hearn L, Derry S, Moore RA. Lacosamide for neuropathic pain and fibromyalgia in adults. Cochrane Database of Systematic Reviews 2012, Issue 2. [DOI: 10.1002/14651858.CD009318.pub2]

Higgins 2003

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

Higgins 2011

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

Häuser 2008

Häuser W, Zimmer C, Felde E, Köllner V. What are the key symptoms of fibromyalgia? Results of a survey of the German Fibromyalgia Association [Was sind die Kernsymptome des Fibromyalgiesyndroms? Umfrageergebnisse der Deutschen Fibromyalgievereinigung]. Schmerz 2008;22(2):176‐83.

Häuser 2010a

Häuser W, Hayo S, Biewer W, Gesmann M, Kühn‐Becker H, Petzke F, et al. Diagnosis of fibromyalgia syndrome ‐ a comparison of Association of the Medical Scientific Societies in Germany, survey, and American College of Rheumatology criteria. Clinical Journal of Pain 2010;26:505‐11.

Häuser 2010b

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:R79.

Häuser 2011a

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 (Hoboken) 2011;63(8):808‐20.

Häuser 2012a

Häuser W, Jung E, Erbslöh‐Möller B, Gesmann M, Kühn‐Becker H, Petermann F, et al. German fibromyalgia consumer reports. Benefits and harms of fibromyalgia syndrome therapies [in German]. Schmerz 2012;26:150‐9.

Häuser 2012b

Häuser W, Sarzi‐Puttini P, Tölle TR, Wolfe F. Placebo and nocebo responses in randomised controlled trials of drugs applying for approval for fibromyalgia syndrome treatment: systematic review and meta‐analysis. Clinical and Experimental Rheumatology 2012;30(6 Suppl 74):78‐87.

Häuser 2013

Häuser W, Urrútia G, Tort S, Üçeyler 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]

Lange 2010

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

Lawrence 2008

Lawrence RC, Felson DT, Helmick CG, Arnold LM, Choi H, Deyo RA, et al. Estimates of the prevalence of arthritis and other rheumatic conditions in the United States. Part II. Arthritis and Rheumatism 2008;58(1):26‐35.

Marschall 2011

Marschall U, Arnold B, Häuser W. Treatment and healthcare costs of fibromyalgia syndrome in Germany: analysis of the data of the Barmer health insurance (BEK) from 2008‐2009 [Behandlung und Krankheitskosten des Fibromyalgie Syndroms in Deutschland]. Schmerz 2011;25(4):402‐10.

Mease 2009

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

Moore 2009

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

Moore 2010a

Moore AR, Eccleston C, Derry S, Wiffen P, Bell RF, Straube S. "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. 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 2011

Moore RA, Wiffen PJ, Derry S, McQuay HJ. Gabapentin for chronic neuropathic pain and fibromyalgia in adults. Cochrane Database of Systematic Reviews 2011, Issue 3. [DOI: 10.1002/14651858.CD007938.pub2]

Moore 2012a

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

Moore 2012b

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.

Mutschler 2011

Mutschler J, Grosshans M, Herwig U, Heekeren K, Kawohl W, Brühl A. Pregabalin‐induced suicidal ideations. Pharmacopsychiatry 2011;44:119.

Nishishinya 2006

Nishishinya MB, Walitt B, Urrútia G, Mease P, Rodríguez A, Lizardo RJR, 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.

Olaizola 2006

Olaizola I, Ellger T, Young P, Bösebeck F, Evers S, Kellinghaus C. Pregabalin‐associated acute psychosis and epileptiform EEG‐changes. Seizure 2006;15:208‐10.

Page 2008

Page RL, Cantu M, Lindenfeld J, Hergott LJ, Lowes BD. Possible heart failure exacerbation associated with pregabalin: case discussion and literature review. Journal of Cardiovascular Medicine (Hagerstown) 2008;9:922‐5.

Phillips 2011

Phillips K, Clauw DJ. Central pain mechanisms in chronic pain states ‐ maybe it is all in their head. Best Practice and Research Clinical Rheumatology 2011;25:141‐54.

RevMan 2012 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.2. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2012.

Routman 2010

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

Seidel 2013

Seidel S, Aigner M, Ossege M, Pernicka E, Wildner B, Sycha T. Antipsychotics for acute and chronic pain in adults. Cochrane Database of Systematic Reviews 2013, Issue 8. [DOI: 10.1002/14651858.CD004844.pub3]

Sommer 2012

Sommer C, Häuser W, Burgmer M, Engelhardt R, Gerhold K, Petzke F, et al. Etiology and pathophysiology of fibromyalgia syndrome. Schmerz 2012;26:259‐67.

Straube 2010

Straube S, Derry S, Moore RA, McQuay HJ. Pregabalin in fibromyalgia: meta‐analysis of efficacy and safety from company clinical trial reports. Rheumatology 2010;49:706‐15.

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]

Tzellos 2010

Tzellos TG, Toulis KA, Goulis DG, Papazisis G, Zampeli VA, Vakfari A, et al. Gabapentin and pregabalin in the treatment of fibromyalgia: a systematic review and a meta‐analysis. Journal of Clinical Pharmacology and Therapeutics 2010;35:639‐56.

Ulloa 2009

Ulloa CM, Towfigh A, Safdieh J. Review of levetiracetam, with a focus on the extended release formulation, as adjuvant therapy in controlling partial‐onset seizures. Neuropsychiatric Disease and Treatment 2009;5:467‐76.

Wiffen 2013a

Wiffen PJ, Derry S, Moore R, Aldington D, Cole P, Rice ASC, et al. Antiepileptic drugs for neuropathic pain and fibromyalgia. Cochrane Database of Systematic Reviews 2013, Issue 6. [DOI: 10.1002/14651858.CD010567]

Wiffen 2013b

Wiffen PJ, Derry S, Lunn MPT, Moore RA. Topiramate for neuropathic pain and fibromyalgia in adults. Cochrane Database of Systematic Reviews 2013, Issue 8. [DOI: 10.1002/14651858.CD008314.pub3]

Winkelmann 2011

Winkelmann A, Perrot S, Schaefer C, Ryan K, Chandran A, Sadosky A, et al. Impact of fibromyalgia severity on health economic costs: results from a European cross‐sectional study. Applied Health Economics and Health Policy 2011;9(2):125‐36.

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 1997

Wolfe F, Anderson J, Harkness D, Bennett RM, Caro XJ, Goldenberg DL, et al. A prospective, longitudinal, multicenter study of service utilization and costs in fibromyalgia. Arthritis and Rheumatism 1997;40(9):1553‐5.

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 2011

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 2011a

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 and Research (Hoboken) 2013;65:777‐85.

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 Pain 2013;17:581‐6.

Üçeyler 2013

Üçeyler N, Zeller D, Kahn AK, Kewenig S, Kittel‐Schneider S, Schmid A, et al. Small fibre pathology in patients with fibromyalgia syndrome. Brain 2013;136:1857‐67.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Jump to:

Arnold 2007

Methods

Study setting: Multicentre study with 3 outpatient research centres in the USA

Study design: Parallel

Duration therapy: 12 weeks

Follow‐up: Not performed

Analysis: ITT; LOCF; for the primary analysis of continuous variables collected at more than 2 time points, a longitudinal analysis that compared the rate of change of the outcome during the treatment period between groups was used. The difference in rate of change was estimated by random regression methods, as described elsewhere. A model for the mean of the outcome variable that included terms for treatment, time, treatment‐by‐time interaction and centre was used. Time was modelled as a continuous variable. To account for the correlation of observations among participants, the SAS procedure MIXED (SAS Institute, Cary, NC) with the best fitting of the following covariance structures was used: unstructured, first‐order heterogeneous autoregressive and first‐order autoregressive. The longitudinal analyses used all available observations from all time points from all participants who completed a baseline evaluation. As a secondary analysis, changes from baseline to end point (the LOCF method) were analysed using an analysis of variance model, with a term for centre

Participants

Participants: 150 (90% women, 97% white, mean age 48 years)

Inclusion criteria: ACR 1990 criteria; age ≥ 18 years; participants were required to score 4 on the mean pain severity item of the BPI

Exclusion criteria: Pain from traumatic injury or structural or regional rheumatic disease; rheumatoid arthritis, inflammatory arthritis or autoimmune disease; unstable medical or psychiatric illness; lifetime history of psychosis, hypomania or mania, epilepsy or dementia; substance abuse in the last 6 months; serious risk of suicide; pregnancy or breastfeeding; unacceptable contraception in those of childbearing potential; participants who, in the opinion of the investigator, were treatment refractory; prior treatment with gabapentin or pregabalin; and treatment with an investigational drug within 30 days of screening. Concomitant medication exclusions consisted of medications or herbal agents with CNS effects, with the exception of episodic use of sedating antihistamines (antidepressants required a 14‐day washout period prior to beginning study medication except for fluoxetine, which required a 30‐day washout period); analgesics, with the exception of paracetamol (acetaminophen) or over‐the‐counter NSAIDs; and unconventional or alternative therapies

Interventions

Active drug: Gabapentin flexible 1200‐2400 mg/d (75 participants): 300 mg once a day at bedtime for 1 week, 300 mg twice a day for 1 week, 300 mg twice a day and 600 mg once a day at bedtime for 2 weeks, 600 mg 3 times a day for 2 weeks, and 600 mg twice a day and 1200 mg once a day at bedtime (2400 mg/d) for the remainder of the study beginning at week 6. If a participant could not tolerate 2400 mg/d, the dosage was reduced to a minimum of 1200 mg/d, administered 3 times a day. The study medication dose was stable for at least the last 4 weeks of the therapy phase. During the tapering phase, the dosage was decreased by 300 mg/d until discontinuation

Placebo: 75 participants

Rescue or allowed medication, or both: Paracetamol (acetaminophen) or over‐the‐counter NSAIDs (dosage not reported)

Outcomes

Pain: BPI mean pain severity (NRS 0‐10). LOCF analysis reported; 50% pain reduction rates not reported and calculated by imputation method

Fatigue: FIQ (VAS 0‐10): Not reported

Sleep: BPI sleep interference (NRS 0‐10). Observed cases reported

Depression: Montgomery Asberg Depression Rating Scale score (NRS 0‐60). Observed cases reported

Anxiety: FIQ (VAS 0‐10): Not reported

Disability: BPI interference from pain (NRS 0‐10). Observed cases reported

Quality of life: FIQ total score (0‐80). Observed cases reported

Patient‐perceived improvement: PGIC: Data extracted from figure

AEs: At the randomisation visit, and at each subsequent visit until the end of the therapy phase, AEs were reviewed (no details reported)

Notes

Safety: Gabapentin‐treated participants reported dizziness, sedation, lightheadedness and weight gain significantly more frequently than did placebo‐treated participants. Most treatment‐emergent AEs were mild to moderate in severity. There were no clinically important findings in the laboratory results, physical examinations, or ECGs

Funding sources and any declaration of interest of primary investigators: Supported by NIH grant N01‐AR‐2‐2264 from the National Institute of Arthritis and Musculoskeletal and Skin Diseases (Dr. Arnold, principal investigator). Dr. Arnold received consulting fees from Eli Lilly (more than USD10,000) and from Pfizer, Cypress Bioscience, Wyeth Pharmaceuticals, Sanofi‐Aventis, Böehringer Ingelheim, Sepracor, Forest Laboratories, Allergan, and Vivus (< USD10,000 each). She also has received research support from Eli Lilly, Pfizer, Cypress Bioscience, Wyeth Pharmaceuticals, Sanofi‐Aventis and Böehringer Ingelheim. Dr. Keck received consulting fees (< USD10,000) from, or is a member of the scientific advisory boards of, Abbott, AstraZeneca Pharmaceuticals, Bristol‐Myers Squibb, GlaxoSmithKline, Eli Lilly and Pfizer. He is a principal or co‐investigator on research studies sponsored by Abbott, the American Diabetes Association, AstraZeneca Pharmaceuticals, Bristol‐Myers Squibb, GlaxoSmithKline, Eli Lilly, Janssen Pharmaceutica, the National Institute of Mental Health, the National Institute of Drug Abuse, Pfizer, the Stanley Medical Research Institute and UCB

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 (details provided 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)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Independent data imputation and statistical analysis (details reported on request)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Observed cases analysis

Selective reporting (reporting bias)

High risk

Outcomes anxiety and fatigue not reported

Arnold 2008

Methods

Study setting: Multicentre study with 84 outpatient research centres in the USA

Study design: Parallel

Duration therapy: 14 weeks

Follow‐up: Not performed

Analysis: ITT; LOCF; analysis of covariance with treatment centre and baseline scores as covariates

Participants

Participants: 745 (95% women, 91% white, mean age 50 years)

Inclusion criteria: ACR 1990 criteria; age ≥ 18 years; pain score of at least 40 on a 100 VAS

Exclusion criteria: Inflammatory rheumatic diseases; active infections; untreated endocrine disorder; severe painful disorder (e.g. painful diabetic peripheral neuropathy, postherpetic pain) that might confound the assessment of pain due to FM; unstable medical or psychiatric disorder (e.g. serious hepatic, respiratory, neurological, haematological or immunological illness, unstable cardiovascular disease; or any other severe acute or chronic medical or psychiatric condition or laboratory abnormalities, including creatinine clearance < 50 mL/min; history of illicit alcohol or drug abuse within the past 2 years; pending workers's compensation, current receipt of disability; past or pending litigation for monetary compensation related to FM; other concomitant medication for FM (antidepressants, anticonvulsants) as well as agents used to treat pain or insomnia

Interventions

Active drug: Pregabalin 300 mg/d (183 participants), pregabalin 450 mg/d (190 participants), pregabalin 600 mg/d (188 participants) twice/d, 2 weeks' dose escalation

Placebo: 184 participants

Rescue or allowed medication: Paracetamol (acetaminophen) < 4 g/d, aspirin < 325 mg/d

Outcomes

Pain: Daily diary mean pain (NRS 0‐10)

Fatigue: MAF (NRS 1‐50)

Sleep: MOS Sleep Problems Index (NRS 0‐100)

Depression: HADS (NRS 0‐21)

Anxiety: HADS (NRS 0‐21)

Disability: SF‐36, physical functioning (NRS 50‐0)

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

Patient‐perceived improvement: PGIC (1‐7)

AEs: Observed or spontaneously reported AEs; laboratory results, physical examinations, ECG

Notes

Safety: Of the 745 participants who received study medication, 614 (82%) had at least 1 AE and 473 (53%) had at least 1 AE that was judged to be related with treatment. The incidence of all‐causality AEs was higher with pregabalin (72%) than with placebo (38%). The severity of the majority of treatment‐related AEs was mild to moderate (89% pregabalin vs. 98% placebo)

Dizziness and somnolence were the most common AEs in pregabalin groups leading to discontinuation of treatment. There were no clinically important findings in the laboratory results, physical examinations or ECGs

Funding sources and any declaration of interest of primary investigators: Dr. Arnold received consulting fees from Eli Lilly, Pfizer, Cypress Biosciences, Wyeth Pharmaceuticals, Sanofi‐Aventis, Böehringer Ingelheim, Allergan, Forest Laboratories and Vivus. Dr. Arnold received research grant support from Eli Lilly, Pfizer, Cypress Biosciences, Wyeth Pharmaceuticals, Sanofi‐Aventis, Böehringer Ingelheim, Allergan and Forest Laboratories. Dr. Arnold is on the Speakers Bureau for Eli Lilly and Pfizer. Dr. Russell has consulted for conducted research studies for Pfizer, Autoimmune Technologies, LLC, LKB World, Jazz Pharmaceuticals, Gruenethal and Allergan. He is on speaker's panel for Merck, Ortho‐McNeil and Pfizer. Erdal Diri received research grants from Hoffmann‐LaRoche, Pfizer, Pain Therapeutics, Proctor & Gamble Pharmaceutics and Corona. He is a speaker and consultant for Pfizer, Amgen, Centecor and Abbott. Rachel Duan, James Youg, Susan Martin, Jeannette Barreite and George Haig are employees of Pfizer and own Pfizer stocks. Uma Sharma is a consultant for Pfizer, Wyeth, Eisal, Analgesic Research and Amgen. Editorial support was provided by Yilmarie Yanchik, an employee of Pfizer. Statistical support was provided by Ed Whalen, an employee of Pfizer

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 (details provided 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)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Independent data imputation and statistical analysis (details reported on request)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Imputation using LOCF and ITT analysis for efficacy data

Selective reporting (reporting bias)

Low risk

All outcomes reported

Crofford 2005

Methods

Study setting: Multicentre study with 40 outpatient research centres in the USA

Study design: Parallel

Duration therapy: 8 weeks

Follow‐up: Not performed

Analysis: ITT; LOCF; analysis of covariance, with treatment and centre as the main effects and the baseline value as the covariate

Participants

Participants: 529 (92% women, 93% white, mean age 49 years)

Inclusion criteria: ACR 1990 criteria; age ≥ 18 years; score of 40 mm on the 100‐mm VAS of the SF‐MPQ

Exclusion criteria:

Inflammatory rheumatic disease or other severe painful disorders that might confound assessment of FM pain. People were also excluded if they had clinically significant or unstable medical or psychological conditions that, in the opinion of the investigator, would compromise participation in the study. Participants with a calculated creatinine clearance rate of 60 mL/min (Cockroft‐Gault equation) were specifically excluded. Those who had failed to respond to previous treatment with gabapentin at dosages of 1200 mg/d for pain associated with FM were excluded. Women who were not postmenopausal were tested to confirm that they were not pregnant or breastfeeding during the study, and all women of childbearing potential were advised to use contraception reliably. Participants who were receiving disability, applying for disability, or engaged in litigation related to FM were excluded

Interventions

Active drug: Pregabalin 150 mg/d (132 participants), pregabalin 300 mg/d (134 participants), pregabalin 450 mg/d (132 participants) 3 times/d

Placebo: 131 participants

Rescue or allowed medication: Paracetamol (acetaminophen) < 4 g/d, aspirin < 325 mg/d

Outcomes

Pain: SF‐MPQ (VAS 0‐100)

Fatigue: MAF (NRS 1‐50)

Sleep: MOS Sleep Problems Index (NRS 0‐100)

Depression: HADS (NRS 0‐21): details provided on request

Anxiety: HADS (NRS 0‐21); details provided on request

Disability: SF‐36, physical functioning (NRS 50‐0)

Quality of life: Not assessed

Patient‐perceived improvement: PGIC: not reported

AEs: All spontaneously reported or observed treatment‐emergent AEs were recorded at each clinic visit, along with the dates on which they began and ended. The sponsor classified AEs using Coding Symbols for a Thesaurus of Adverse Reaction Terms, 4th Edition (COSTART IV) . A 12‐lead ECG was recorded at screening and end point, and clinical laboratory tests (haematology, urinalysis and chemistry) were performed at screening, week 3, and end point. Serious AE not reported

Notes

Safety: Most AEs were mild or moderate. Dizziness and somnolence were the 2 most frequently reported AEs and tended to be dose‐related across pregabalin groups. Non‐CNS AEs that were more frequent in the pregabalin groups included weight gain and peripheral oedema. There were no clinically important findings in the analyses of haematology, blood chemistry or urinalysis. Similarly, there were no clinically significant findings in the visual function, physical, or neurological examinations or on the ECGs

Funding sources and any declaration of interest of primary investigators: Supported by Pfizer Global Research and Development, Ann
Arbor, Michigan. Dr. Crofford has received consulting fees of < USD10,000 from Cypress Bioscience, Eli Lilly & Co, Orphan Pharmaceuticals, Pfizer and Wyeth. Dr. Rowbotham has received consulting fees of < USD10,000 from Eli Lilly & Co and Xenoport, owns stock in Xenoport and Neuromolecular and was a co‐investigator on a study of gabapentin funded by Pfizer. Dr. Mease has received consulting fees of < USD10,000 from Pfizer, Cypress Bioscience, Eli Lilly & Co and Pierre Fabre and owns stock in Cypress Bioscience. Dr. Dworkin has served on the advisory board or as a consultant for fees of < USD10,000 for Abbott Laboratories, Alpharma, AstraZeneca Pharmaceuticals, Bristol‐Myers Squibb, Elan Pharmaceuticals, Eli Lilly & Co, GlaxoSmithKline, Johnson & Johnson, Merck KGaA, NeurogesX Inc., Ortho‐McNeil Pharmaceutical and UCB Pharma; has received consulting fees of > USD10,000 from Pfizer, Allergan, Novartis, Epicept and Endo; and owns stock in NeurogesX, Inc. Ms Corbin, Mr. Young, Ms LaMoreaux, Ms Martin and Dr. Sharma own stock in Pfizer

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 (details provided 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

Independent data imputation and statistical analysis (details reported on request)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Imputation using LOCF and ITT analysis for efficacy data

Selective reporting (reporting bias)

High risk

Quality of life scores and SAEs not reported and not provided on request

Mease 2008

Methods

Study setting: Multicentre study with 79 outpatient research centres in the USA

Study design: Parallel

Duration therapy: 13 weeks

Follow‐up: Not performed

Analysis: ITT; LOCF; analysis of covariance with treatment centre and baseline scores as covariates

Participants

Participants: 558 (94% women, 91% white, mean age 49 years)

Inclusion criteria: ACR 1990 criteria; age ≥ 18 years; pain score of at least 4 on a 0‐10 NRS

Exclusion criteria: Inflammatory or rheumatological disease; other severe pain disorders; clinically significant or unstable medical or psychological conditions; creatinine clearance < 60 mL/min, severe depression, receiving or applying for disability benefits

Interventions

Active drug: pregabalin 300 mg/d (185 participants), pregabalin 450 mg/d (183 participants), pregabalin 600 mg/d (190 participants) twice/d, 2 weeks' dose escalation

Placebo: 190 participants

Rescue or allowed medication: paracetamol (acetaminophen) < 4 g/d

Outcomes

Pain: Daily diary mean pain (NRS 0‐10); 50% pain reduction rates provided on request

Fatigue: MAF (NRS 1‐50), details provided on request

Sleep: MOS Sleep Problems Index (NRS 0‐100)

Depression: HADS (NRS 0‐21): details provided on request

Anxiety: HADS (NRS 0‐21): details provided on request

Disability: SF‐36, physical functioning (NRS 50‐0): data provided on request

Quality of life: FIQ total score (0‐100); standard deviations provided on request

Patient‐perceived improvement: PGIC (1‐7)

AEs: Volunteered by participants or observed by the clinician at every visit, laboratory results, physical examinations, ECG

Notes

Safety: At least 1 AE was reported by 89.2% of participants with pregabalin 300 mg/d, 91.8% of participants with pregabalin 450 mg/d, 93.7% of participants with pregabalin 600 mg/d and by 76.3% of participants with placebo

Dizziness, somnolence and weight gain were more common in pregabalin groups compared with placebo. There were no clinically important findings in the laboratory results, physical examinations, or ECGs

Funding sources and any declaration of interest of primary investigators: Supported by Pfizer Global Research and Development, Ann
Arbor, Michigan. Dr. Mease has received research grant support from Pfizer, Cypress Bioscience, Forest Laboratories, Eli Lilly, Allergan, Wyeth Pharmaceuticals, Jazz Pharmaceuticals and Fralex Therapeutics. Dr. Russell has received research grant support from Pfizer, Eli Lilly, Allergan, Böehringer Ingelheim, Cypress Bioscience, Jazz Pharmaceuticals, Grünenthal and Pierre Fabre. Dr. Arnold has received research grant support from Eli Lilly, Pfizer, Cypress Biosciences, Wyeth Pharmaceuticals, Sanofi‐Aventis, Böehringer Ingelheim, Allergan and Forest Laboratories. Dr. Florian, Mr. Young and Ms Mareen are employees of Pfizer and own stock of Pfizer. Dr Sharma was an employee of Pfizer when the study was conducted and owns stocks of Pfizer

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 reported on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Independent data imputation and statistical analysis (details reported on request)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Imputation using LOCF and ITT analysis for efficacy data

Selective reporting (reporting bias)

High risk

Disability not reported

Ohta 2012

Methods

Study setting: Multicentre study with 44 outpatient research centres in Japan

Study design: Parallel

Duration therapy: 14 weeks

Follow‐up: Not performed

Analysis: ITT; LOCF; a mixed‐effect model taking baseline value as covariate was used for the analysis, which included participants as the random effect and dose groups, points at time of evaluation, and interaction between a dose group and its point at time of evaluation as the fixed effects

Participants

Participants: 501 (89% women, 10% Japanese, mean age 47 years)

Inclusion criteria: ACR 1990 criteria; age ≥ 18 years; pain score of ≥ 40 mm on the 100‐mm VAS at visit 2, and had assessed and documented their pain score on at least 4
of the past 7 days prior to visit 2 while recording a mean pain score of ≥ 4 on the 11‐point NRS

Exclusion criteria: Decrease of ≥ 30% on their pain VAS during the placebo run‐in period (at visit 2 compared with visit 1), in order to remove potential placebo‐responders. Participants were also excluded if they were being treated for depression or if they were at risk of suicide or self harm in the opinion of the study investigator; inflammatory or rheumatological disease; other severe pain disorders; clinical significant or unstable medical or psychological conditions; history of malignancy; creatinine clearance < 60 mL/min, severe depression, receiving or applying for disability benefits

Interventions

Active drug: Pregabalin (251 participants): treatment was started at 150 mg/d, escalated to 300 mg/d 1 week later, and to 450 mg/d after another week. The dose was adjusted (increased or decreased) until visit 5 of the study, after which the maintenance dose was either 300 or 450 mg/d

Placebo: 250 participants

Rescue or allowed medication: Paracetamol (acetaminophen) or NSAIDs for additional pain relief, although for NSAIDs, the participant must have already been on a stable regimen for longer than 30 days

Outcomes

Pain: Daily diary mean pain (NRS 0‐10)

Fatigue: FIQ Fatigue single scale (VAS 0‐10)

Sleep: MOS Sleep Problems Index (NRS 100‐0)

Depression: HADS (NRS 0‐21)

Anxiety: HADS (NRS 0‐21)

Disability: SF‐36, physical functioning (NRS 50‐0)

Quality of life: FIQ total score (0‐100)

Patient‐perceived improvement: PGIC (1‐7)

AEs: volunteered by participants or observed by the clinician at every visit, laboratory results, physical examinations, ECG

Notes

Safety: The incidence of all‐causality AEs was higher with pregabalin (occurring in 225 of 250 participants, 90.0%) than with placebo (175 of 248 participants, 70.6%). Similarly, the incidence of treatment‐related AEs was higher with pregabalin (206 of 250 participants, 82.4%) than with placebo (128 of 248 participants, 51.6%). The most common AEs in this study were somnolence, dizziness, nasopharyngitis, increased weight and constipation with pregabalin treatment, and somnolence and nasopharyngitis with placebo.

A laboratory test result of increased creatine kinase was more frequent with pregabalin (7 of 250 participants, 2.8%) than with placebo (1 of 248 participants, 0.4%), although all cases were of mild severity. Increased weight was reported more frequently with pregabalin (39 of 250 participants, 15.6% (38 mild, 1 moderate)) than with placebo (9 of 248 participants, 3.6% (8 mild, 1 moderate)). There were no clinically significant changes in blood pressure or pulse rate in the pregabalin and placebo group.
There were 4 serious AEs in 4 (0.8%) participants who had received ≥ 1 dose of treatment; 1 participant was from the placebo group (abnormal liver function test result) and 3 were from the pregabalin group (breast cancer, viral gastroenteritis and musculoskeletal stiffness). Severe AEs were observed in 2 participants from the pregabalin group (breast cancer and loss of consciousness, each in 1 participant) and all other AEs were mild or moderate. As assessed by the study investigators, there was no causal relationship to the study drug for any serious or severe AEs.
Suicidal ideation (mild), as rated by C‐SSRS, was noted in 2 participants in the pregabalin group. The investigator (a physician specialising in psychosomatic medicine) had noted suicidal ideation in each of these people prior to the start of the study. In each case, incidence was attributable to family environment and was judged to have no causal relationship to the study drug. The suicidal ideation in these cases was not judged by the study investigator to be a real desire, and treatment with the study drug was continued.

Funding sources and any declaration of interest of primary investigators: The study was funded by Pfizer Japan Inc. Medical writing support was provided by Joshua Fink PhD, of UBC Scientific Solutions, and funded by Pfizer Inc. HOhta, MO and MS are employees of Pfizer Japan, Inc. KN and HOka received a consultancy fee from Pfizer Japan, Inc. for their participation in this study. CU declares no competing interests. KN, HOka and CU were not compensated for their work on the manuscript.

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 reported on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Independent data imputation and statistical analysis (details reported on request)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Imputation using LOCF and ITT analysis for efficacy data

Selective reporting (reporting bias)

Low risk

All outcomes reported on request

Pauer 2011

Methods

Study setting: Multicentre study with 73 outpatient research centres in the USA, Middle America, South America, Western Europe, Asia, Australia and India

Study design: Parallel

Duration therapy: 14 weeks

Follow‐up: Not performed

Analysis: ITT; LOCF; analysis of covariance with treatment centre, week, treatment by week interaction and baseline scores as covariates

Participants

Participants: 986 (91% women, 76% white, mean age 49 years)

Inclusion criteria: ACR 1990 criteria; age ≥ 18 years; pain score of at least 40 on a 100 VAS

Exclusion criteria: Participants who demonstrated a high placebo response (≥ 30% decrease on the VAS following the 1‐week run‐in period compared with screening). Provided on request: other severe pain conditions; any widespread inflammatory musculoskeletal disorder, active infections or untreated endocrine disorders; previous participation in a clinical trial with pregabalin, previous exposure to pregabalin or currently pregabalin for any condition; severe depression according to the judgement of the investigator; serious internal diseases or any other acute or chronic medical or psychiatric condition or laboratory abnormality that may increase the risk associated with trial participation; intake of any experimental drug within 30 days prior to screening; use of prohibited pain/sleep medication (including antidepressants, sedatives, hypnotics, NSAIDs, opioids, muscle relaxants) in the absence of appropriate washout period; pending disability claims or currently receiving monetary compensation pertinent to the patient's FM or co‐morbid diseases

Interventions

Active drug: Pregabalin 300 mg/d (184 participants), pregabalin 450 mg/d (182 participants), pregabalin 600 mg/d (186 participants) twice/d, 2 weeks' dose escalation

Placebo: 184 participants

Rescue or allowed medication: No details reported

Outcomes

Pain: Daily diary mean pain (NRS 0‐10)

Fatigue: MAF (NRS 1‐50)

Sleep: MOS Sleep Problems Index (NRS 0‐100)

Depression: HADS (NRS 0‐21): data available on clinicaltrials.gov/ct2/show/NCT00333866

Anxiety: HADS (NRS 0‐21): data available on clinicaltrials.gov/ct2/show/NCT00333866

Disability: SF‐36, physical functioning (NRS 50‐0): data available on clinicaltrials.gov/ct2/show/NCT00333866

Quality of life: FIQ total score (0‐100)

Patient‐perceived improvement: PGIC (1‐7)

AEs: AE were recorded at each visit. Investigators rated the severity of each AE and its relationship to study drug. Clinical laboratory evaluations, physical examinations, abbreviated neurological examinations, and 12‐lead ECGs were performed at regular intervals

Notes

Safety: Of the 736 participants receiving study medication, 626 (85%) experienced at least 1 AE. The occurrence of AE increased with dosage (73%, 85%, 90% and 92% for placebo, 300, 450 and 600 mg/d pregabalin‐treated participants, respectively).The AEs most frequently reported by pregabalin‐treated participants were dizziness, somnolence, weight gain, headache, peripheral oedema, fatigue and dry mouth. Most AE were rated by investigators as mild or moderate. 18 participants experienced SAE, 4 participants treated with placebo and 14 pregabalin. Only 1 SAE, an incidence of chest pain in 1 person in the 450 mg/d pregabalin group, was considered by the investigator to be related to treatment and the person was withdrawn from the study. 8 other participants experienced an SAE that led to withdrawal from the study and 1 participant experienced an SAE that led to a dose reduction

There were no clinically relevant differences in clinical laboratory evaluations, vital signs, physical examination or ECG findings

Funding sources and any declaration of interest of primary investigators: Supported by Pfizer. No declaration of interest of primary investigators included

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 reported on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Independent data imputation and statistical analysis (details reported on request)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Imputation using LOCF and ITT analysis for efficacy data

Selective reporting (reporting bias)

Low risk

All outcomes reported

Rowbotham 2012

Methods

Study setting: Single‐centre study in the USA, investigator‐initiated

Study design: Parallel

Duration therapy: 8 weeks

Follow‐up: Not performed

Analysis: Completer analysis by mixed effect regressions

Participants

Participants: 60 participants (93% female, 79% white, mean age 49 years)

Inclusion criteria: ACR 1990 criteria; age ≥ 18 years; McGill Pain Questionnaire VAS of at least 40mm on 100‐mm scale at screening and mean daily diary pain at least 4 out of 10 during the 7 days prior to study drug initiation

Exclusion criteria: Pregnant or lactating women, hypersensitivity to levetiracetam or prior treatment with it; clinically significant liver, kidney or haematological disorders, a Westergen erythrocyte sedimentation rate exceeding 40 mm/min, abnormal elevated antinuclear antibody (1:160) or rheumatoid factor (> 80 IU/mL) levels, another explanation for their pain, illicit drug or alcohol abuse within the last year, involvement in unsettled litigation pertaining to their FM (such as automobile accident, civil lawsuit or worker's compensation), and ongoing monetary compensation as a result of litigation or disability claims with the exception of US social security disability benefits; subjects who are considered unreliable as to medication compliance or adherence to scheduled appointments as determined by the investigators; subjects who have serious or unstable medical or psychological conditions that in the opinion of the investigator(s), would compromise the subject's participation in the study

Interventions

Active drug: 40 participants were titrated to a maximum 3000 mg/d over a 6‐week period and tapered off study medication after 8 weeks of treatment

Placebo: 26 participants

Rescue or allowed medication: Continuation of stable doses of antidepressants and opiates allowed

Outcomes

Pain: Diary pain (NRS 0‐10); 30% and 50% pain reduction not reported and calculated by imputation method

Fatigue: FIQ Fatigue single scale not reported

Sleep: Diary sleep interference (NRS 0‐10)

Depression: FIQ Depression not reported

Anxiety: FIQ Anxiety not reported

Disability: FIQ Disability not reported

Quality of life: FIQ total score (0‐100)

Patient‐perceived improvement: PGIC (1‐7)

AEs: Subject‐reported AEs were classified into 1 of 39 categories and a subject was considered positive even if they reported an AE at only 1 visit after beginning treatment

Notes

Safety: There were no SAEs in either group. AEs such as thinking abnormal, restlessness, itching and movement disorder were significantly more frequent in the levetiracetam group

Funding sources and any declaration of interest of primary investigators: Supported by an investigator‐initiated grant from UCB Pharma and NIH grant K24 NS02164. No declaration of interest of primary investigator included

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 reported on request)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Independent data imputation and statistical analysis (details reported on request)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only observed cases data available

Selective reporting (reporting bias)

High risk

Outcomes FIQ subscales fatigue, anxiety, depression, disability not reported

UCB 2006

Methods

Study setting: Multicentre study, number of outpatient research centres in the USA not reported

Study design: Parallel

Duration therapy: 12 weeks

Follow‐up: Not performed

Analysis: ITT; LOCF; analysis of covariance with treatment centre, week, treatment by week interaction and baseline scores as covariates

Participants

Participants: 159 (93% women, race not reported, mean age 50 years)

Inclusion criteria: Not reported

Exclusion criteria: Not reported

Interventions

Active drug: Lacosamide 400 mg (81 participants), 4‐week titration from 100 mg/d to 400 mg/d, increasing by 100 mg/d at weekly intervals; 8‐week maintenance

Placebo: 78 participants

Rescue or allowed medication: No details reported

Outcomes

Pain: Daily diary mean pain (NRS 0‐10)

Fatigue: Fatigue score of FIQ (VAS 0‐10)

Sleep: Mean daily interference with sleep (NRS 0‐10)

Depression: HADS (NRS 0‐21)

Anxiety: HADS (NRS 0‐21)

Disability: Mean daily interference with activity (NRS 0‐10)

Quality of life: FIQ total score (0‐100)

Patient‐perceived improvement: PGIC (1‐7)

AEs: No details reported

Notes

Safety: No deaths in either group. No further details reported

Funding sources and any declaration of interest of primary investigators: Supported by UCB. No declaration of interest of primary investigators included

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not reported

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double blind (number and appearance of placebo capsules similar)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participant reported and participant blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

Imputation using LOCF for continuous data reported, but not used. ITT for PGIC, AEs and withdrawals

Selective reporting (reporting bias)

High risk

No details of AEs reported

ACR: American College of Rheumatology; AE: adverse event; BPI: Brief Pain Inventory; C‐SSRS: Columbia Suicide Severity Rating Scale; CNS: central nervous system; ECG: electrocardiogram; FIQ: Fibromyalgia Impact Questionnaire; FM: fibromyalgia; HADS: Hospital Anxiety and Depression Scale; ITT: intention to treat; LOCF: last observation carried forward; MAF: Multidimensional Assessment of Fatigue; MOS: Medical Outcomes Study; NIH: National Institutes of Health; NRS: numeric rating scale; NSAID: non‐steroidal anti‐inflammatory drug; PGIC: Patient Global Impression of Change; SAE: serious adverse events; SF‐36: Short‐Form Health Survey ‐ 36 items; SF‐MPQ: Short‐Form McGill Pain Questionnaire; VAS: visual analogue scale.

Characteristics of excluded studies [ordered by study ID]

Jump to:

Study

Reason for exclusion

Crofford 2008

Pregabalin: Study design (enriched enrolment randomised withdrawal design) over 26 weeks cannot be combined with parallel or cross‐over designs for meta‐analysis

Roth 2012

Pregabalin: Pregabalin and placebo treatment 4 weeks each within a cross‐over design

Data and analyses

Open in table viewer
Comparison 1. Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain Show forest plot

1

119

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

‐0.49 [‐0.86, ‐0.13]

Analysis 1.1

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 1 Pain.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 1 Pain.

2 50% pain reduction Show forest plot

1

150

Risk Ratio (IV, Random, 95% CI)

1.6 [1.01, 2.53]

Analysis 1.2

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 2 50% pain reduction.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 2 50% pain reduction.

3 Sleep problems Show forest plot

1

119

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

‐0.71 [‐1.08, ‐0.34]

Analysis 1.3

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.

4 Health‐related quality of life Show forest plot

1

119

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

‐0.66 [‐1.03, ‐0.29]

Analysis 1.4

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.

5 Withdrawal due to adverse events Show forest plot

1

150

Risk Ratio (IV, Random, 95% CI)

1.71 [0.71, 4.11]

Analysis 1.5

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 5 Withdrawal due to adverse events.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 5 Withdrawal due to adverse events.

6 Dizziness Show forest plot

1

150

Risk Ratio (IV, Random, 95% CI)

2.71 [1.21, 6.07]

Analysis 1.6

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.

7 30% pain reduction Show forest plot

1

150

Risk Ratio (M‐H, Random, 95% CI)

1.65 [1.10, 2.48]

Analysis 1.7

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 7 30% pain reduction.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 7 30% pain reduction.

8 Depression Show forest plot

1

119

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

‐0.52 [‐0.89, ‐0.16]

Analysis 1.8

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 8 Depression.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 8 Depression.

9 Disability Show forest plot

1

119

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

‐0.94 [‐1.32, ‐0.56]

Analysis 1.9

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 9 Disability.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 9 Disability.

Open in table viewer
Comparison 2. Lacosamide 400 mg/day versus placebo at end of treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain Show forest plot

1

158

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

‐0.25 [‐0.56, 0.07]

Analysis 2.1

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 1 Pain.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 1 Pain.

2 Fatigue Show forest plot

1

121

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

‐0.07 [‐0.43, 0.28]

Analysis 2.2

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 2 Fatigue.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 2 Fatigue.

3 Sleep problems Show forest plot

1

158

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

‐0.19 [‐0.51, 0.12]

Analysis 2.3

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.

4 Health‐related quality of life Show forest plot

1

157

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

‐0.15 [‐0.47, 0.16]

Analysis 2.4

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.

5 Serious adverse events Show forest plot

1

159

Risk Ratio (IV, Random, 95% CI)

0.15 [0.01, 2.82]

Analysis 2.5

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 5 Serious adverse events.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 5 Serious adverse events.

6 Dizziness Show forest plot

1

159

Risk Ratio (IV, Random, 95% CI)

2.34 [1.08, 5.06]

Analysis 2.6

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.

7 Anxiety Show forest plot

1

133

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

0.0 [‐0.34, 0.34]

Analysis 2.7

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 7 Anxiety.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 7 Anxiety.

8 Depression Show forest plot

1

134

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

0.11 [‐0.23, 0.45]

Analysis 2.8

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 8 Depression.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 8 Depression.

9 Disability Show forest plot

1

158

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

‐0.19 [‐0.51, 0.12]

Analysis 2.9

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 9 Disability.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 9 Disability.

10 Patient Global Impression of Change 'much' or 'very much' improved Show forest plot

1

134

Risk Ratio (IV, Random, 95% CI)

1.32 [0.85, 2.04]

Analysis 2.10

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 10 Patient Global Impression of Change 'much' or 'very much' improved.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 10 Patient Global Impression of Change 'much' or 'very much' improved.

Open in table viewer
Comparison 3. Levetiracetam up to 3000 mg/day versus placebo at end of treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain Show forest plot

1

50

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

‐0.21 [‐0.77, 0.36]

Analysis 3.1

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 1 Pain.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 1 Pain.

2 50% pain reduction Show forest plot

1

66

Risk Ratio (IV, Random, 95% CI)

1.52 [0.43, 5.34]

Analysis 3.2

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 2 50% pain reduction.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 2 50% pain reduction.

3 Sleep problems Show forest plot

1

50

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

‐0.16 [‐0.72, 0.40]

Analysis 3.3

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.

4 Health‐related quality of life Show forest plot

1

50

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

0.14 [‐0.42, 0.70]

Analysis 3.4

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.

5 Withdrawal due to adverse events Show forest plot

1

66

Risk Ratio (IV, Random, 95% CI)

0.87 [0.21, 3.56]

Analysis 3.5

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 5 Withdrawal due to adverse events.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 5 Withdrawal due to adverse events.

6 Dizziness Show forest plot

1

66

Risk Ratio (IV, Random, 95% CI)

1.3 [1.02, 1.66]

Analysis 3.6

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.

7 30% pain reduction Show forest plot

1

66

Risk Ratio (IV, Random, 95% CI)

1.23 [0.65, 2.33]

Analysis 3.7

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 7 30% pain reduction.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 7 30% pain reduction.

Open in table viewer
Comparison 4. Pregabalin versus placebo at end of treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain Show forest plot

5

3252

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

‐0.28 [‐0.35, ‐0.20]

Analysis 4.1

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 1 Pain.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 1 Pain.

1.1 Pregabalin 150 mg/d

1

175

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

‐0.10 [‐0.45, 0.24]

1.2 Pregabalin 300 mg/d

4

913

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

‐0.24 [‐0.39, ‐0.09]

1.3 Pregabalin 450 mg/d

4

915

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

‐0.36 [‐0.51, ‐0.20]

1.4 Pregabalin 600 mg/d

3

751

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

‐0.30 [‐0.51, ‐0.09]

1.5 Pregabalin flexible 300 or 450 mg/d

1

498

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

‐0.24 [‐0.41, ‐0.06]

2 50% pain reduction Show forest plot

5

3256

Risk Ratio (IV, Random, 95% CI)

1.59 [1.33, 1.90]

Analysis 4.2

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 2 50% pain reduction.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 2 50% pain reduction.

2.1 Pregabalin 150 mg/d

1

175

Risk Ratio (IV, Random, 95% CI)

0.92 [0.39, 2.19]

2.2 Pregabalin 300 mg/d

4

915

Risk Ratio (IV, Random, 95% CI)

1.45 [1.03, 2.05]

2.3 Pregabalin 450 mg/d

4

916

Risk Ratio (IV, Random, 95% CI)

1.75 [1.23, 2.49]

2.4 Pregabalin 600 mg/d

3

752

Risk Ratio (IV, Random, 95% CI)

1.51 [1.04, 2.20]

2.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

Risk Ratio (IV, Random, 95% CI)

1.88 [1.26, 2.83]

3 Fatigue Show forest plot

5

3195

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

‐0.17 [‐0.25, ‐0.09]

Analysis 4.3

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 3 Fatigue.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 3 Fatigue.

3.1 Pregabalin 150 mg/d

1

165

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

‐0.20 [‐0.56, 0.15]

3.2 Pregabalin 300 mg/d

4

892

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

‐0.16 [‐0.31, ‐0.01]

3.3 Pregabalin 450 mg/d

4

897

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

‐0.15 [‐0.30, 0.01]

3.4 Pregabalin 600 mg/d

3

743

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

‐0.08 [‐0.25, 0.09]

3.5 Pregabalin flexible 300 or 450 mg/d

1

498

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

‐0.31 [‐0.49, ‐0.14]

4 Sleep problems Show forest plot

5

3193

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

‐0.35 [‐0.43, ‐0.27]

Analysis 4.4

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 4 Sleep problems.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 4 Sleep problems.

4.1 Pregabalin 150 mg/d

1

163

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

‐0.44 [‐0.80, ‐0.08]

4.2 Pregabalin 300 mg/d

4

897

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

‐0.29 [‐0.44, ‐0.14]

4.3 Pregabalin 450 mg/d

4

893

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

‐0.45 [‐0.63, ‐0.27]

4.4 Pregabalin 600 mg/d

3

744

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

‐0.40 [‐0.57, ‐0.23]

4.5 Pregabalin flexible 300 or 450 mg/d

1

496

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

‐0.20 [‐0.37, ‐0.02]

5 Health‐related quality of life Show forest plot

4

2724

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

‐0.17 [‐0.26, ‐0.09]

Analysis 4.5

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 5 Health‐related quality of life.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 5 Health‐related quality of life.

5.1 Pregabalin 300 mg/d

3

738

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

‐0.12 [‐0.28, 0.05]

5.2 Pregabalin 450 mg/d

3

737

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

‐0.24 [‐0.41, ‐0.07]

5.3 Pregabalin 600 mg/d

3

751

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

‐0.14 [‐0.31, 0.02]

5.4 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

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

‐0.20 [‐0.37, ‐0.02]

6 Withdrawal due to adverse events Show forest plot

5

3259

Risk Ratio (IV, Random, 95% CI)

1.68 [1.36, 2.07]

Analysis 4.6

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 6 Withdrawal due to adverse events.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 6 Withdrawal due to adverse events.

6.1 Pregabalin 150 mg/d

1

175

Risk Ratio (IV, Random, 95% CI)

1.19 [0.35, 4.08]

6.2 Pregabalin 300 mg/d

4

917

Risk Ratio (IV, Random, 95% CI)

1.54 [1.02, 2.34]

6.3 Pregabalin 450 mg/d

4

917

Risk Ratio (IV, Random, 95% CI)

2.02 [1.32, 3.09]

6.4 Pregabalin 600 mg/d

3

752

Risk Ratio (IV, Random, 95% CI)

2.53 [1.65, 3.86]

6.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

Risk Ratio (IV, Random, 95% CI)

1.01 [0.65, 1.57]

7 Serious adverse events Show forest plot

4

2729

Risk Ratio (IV, Random, 95% CI)

1.03 [0.71, 1.49]

Analysis 4.7

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 7 Serious adverse events.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 7 Serious adverse events.

7.1 Pregabalin 300 mg/d

3

738

Risk Ratio (IV, Random, 95% CI)

0.96 [0.50, 1.86]

7.2 Pregabalin 450 mg/d

3

741

Risk Ratio (IV, Random, 95% CI)

1.03 [0.52, 2.03]

7.3 Pregabalin 600 mg/d

3

752

Risk Ratio (IV, Random, 95% CI)

1.01 [0.55, 1.87]

7.4 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

Risk Ratio (IV, Random, 95% CI)

2.98 [0.31, 28.42]

8 Dizziness Show forest plot

5

3257

Risk Ratio (IV, Random, 95% CI)

3.77 [3.06, 4.63]

Analysis 4.8

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 8 Dizziness.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 8 Dizziness.

8.1 Pregabalin 150 mg/d

1

175

Risk Ratio (IV, Random, 95% CI)

2.44 [0.91, 6.54]

8.2 Pregabalin 300 mg/d

4

916

Risk Ratio (IV, Random, 95% CI)

3.11 [2.09, 4.65]

8.3 Pregabalin 450 mg/d

4

917

Risk Ratio (IV, Random, 95% CI)

3.95 [2.68, 5.82]

8.4 Pregabalin 600 mg/d

3

751

Risk Ratio (IV, Random, 95% CI)

4.00 [2.65, 6.03]

8.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

Risk Ratio (IV, Random, 95% CI)

4.89 [2.89, 8.28]

9 30% pain reduction Show forest plot

5

3259

Risk Ratio (IV, Random, 95% CI)

1.37 [1.22, 1.53]

Analysis 4.9

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 9 30% pain reduction.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 9 30% pain reduction.

9.1 Pregabalin 150 mg/d

1

176

Risk Ratio (IV, Random, 95% CI)

1.05 [0.62, 1.77]

9.2 Pregabalin 300 mg/d

4

916

Risk Ratio (IV, Random, 95% CI)

1.35 [1.08, 1.68]

9.3 Pregabalin 450 mg/d

4

917

Risk Ratio (IV, Random, 95% CI)

1.49 [1.20, 1.85]

9.4 Pregabalin 600 mg/d

3

752

Risk Ratio (IV, Random, 95% CI)

1.37 [1.07, 1.76]

9.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

Risk Ratio (IV, Random, 95% CI)

1.32 [1.04, 1.68]

10 Anxiety Show forest plot

5

3214

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

‐0.12 [‐0.20, ‐0.04]

Analysis 4.10

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 10 Anxiety.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 10 Anxiety.

10.1 Pregabalin 150 mg/d

1

166

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

0.07 [‐0.28, 0.42]

10.2 Pregabalin 300 mg/d

4

903

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

‐0.05 [‐0.20, 0.10]

10.3 Pregabalin 450 mg/d

4

900

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

‐0.15 [‐0.30, 0.00]

10.4 Pregabalin 600 mg/d

3

749

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

‐0.16 [‐0.32, 0.01]

10.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

496

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

‐0.17 [‐0.35, 0.01]

11 Depression Show forest plot

5

3212

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

‐0.09 [‐0.16, ‐0.01]

Analysis 4.11

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 11 Depression.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 11 Depression.

11.1 Pregabalin 150 mg/d

1

165

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

‐0.10 [‐0.45, 0.25]

11.2 Pregabalin 300 mg/d

4

902

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

‐0.05 [‐0.20, 0.10]

11.3 Pregabalin 450 mg/d

4

900

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

‐0.13 [‐0.28, 0.02]

11.4 Pregabalin 600 mg/d

3

749

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

‐0.06 [‐0.23, 0.10]

11.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

496

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

‐0.09 [‐0.27, 0.08]

12 Disability Show forest plot

5

3145

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

‐0.01 [‐0.11, 0.09]

Analysis 4.12

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 12 Disability.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 12 Disability.

12.1 Pregabalin 150 mg/d

1

166

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

0.11 [‐0.24, 0.47]

12.2 Pregabalin 300 mg/d

4

879

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

0.04 [‐0.11, 0.20]

12.3 Pregabalin 450 mg/d

4

888

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

0.00 [‐0.17, 0.18]

12.4 Pregabalin 600 mg/d

3

714

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

0.03 [‐0.14, 0.21]

12.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

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

‐0.29 [‐0.47, ‐0.12]

13 Patient Global Impression of Change 'much' or 'very much' improved Show forest plot

5

3183

Risk Ratio (IV, Random, 95% CI)

1.38 [1.23, 1.55]

Analysis 4.13

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 13 Patient Global Impression of Change 'much' or 'very much' improved.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 13 Patient Global Impression of Change 'much' or 'very much' improved.

13.1 Pregabalin 150 mg/d

1

175

Risk Ratio (IV, Random, 95% CI)

1.30 [0.74, 2.29]

13.2 Pregabalin 300 mg/d

4

886

Risk Ratio (IV, Random, 95% CI)

1.25 [0.99, 1.57]

13.3 Pregabalin 450 mg/d

4

892

Risk Ratio (IV, Random, 95% CI)

1.54 [1.15, 2.06]

13.4 Pregabalin 600 mg/d

3

734

Risk Ratio (IV, Random, 95% CI)

1.37 [1.06, 1.76]

13.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

496

Risk Ratio (IV, Random, 95% CI)

1.44 [1.11, 1.87]

Study flow diagram.
Figures and Tables -
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.
Figures and Tables -
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.
Figures and Tables -
Figure 3

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

Forest plot of comparison: 4 Pregabalin versus placebo at end of treatment, outcome: 4.2 50% pain reduction.
Figures and Tables -
Figure 4

Forest plot of comparison: 4 Pregabalin versus placebo at end of treatment, outcome: 4.2 50% pain reduction.

Forest plot of comparison: 4 Pregabalin versus placebo at end of treatment, outcome: 4.3 Fatigue.
Figures and Tables -
Figure 5

Forest plot of comparison: 4 Pregabalin versus placebo at end of treatment, outcome: 4.3 Fatigue.

Forest plot of comparison: 4 Pregabalin versus placebo at end of treatment, outcome: 4.4 Sleep problems.
Figures and Tables -
Figure 6

Forest plot of comparison: 4 Pregabalin versus placebo at end of treatment, outcome: 4.4 Sleep problems.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 1 Pain.
Figures and Tables -
Analysis 1.1

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 1 Pain.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 2 50% pain reduction.
Figures and Tables -
Analysis 1.2

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 2 50% pain reduction.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.
Figures and Tables -
Analysis 1.3

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.
Figures and Tables -
Analysis 1.4

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 5 Withdrawal due to adverse events.
Figures and Tables -
Analysis 1.5

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 5 Withdrawal due to adverse events.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.
Figures and Tables -
Analysis 1.6

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 7 30% pain reduction.
Figures and Tables -
Analysis 1.7

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 7 30% pain reduction.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 8 Depression.
Figures and Tables -
Analysis 1.8

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 8 Depression.

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 9 Disability.
Figures and Tables -
Analysis 1.9

Comparison 1 Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment, Outcome 9 Disability.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 1 Pain.
Figures and Tables -
Analysis 2.1

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 1 Pain.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 2 Fatigue.
Figures and Tables -
Analysis 2.2

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 2 Fatigue.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.
Figures and Tables -
Analysis 2.3

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.
Figures and Tables -
Analysis 2.4

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 5 Serious adverse events.
Figures and Tables -
Analysis 2.5

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 5 Serious adverse events.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.
Figures and Tables -
Analysis 2.6

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 7 Anxiety.
Figures and Tables -
Analysis 2.7

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 7 Anxiety.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 8 Depression.
Figures and Tables -
Analysis 2.8

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 8 Depression.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 9 Disability.
Figures and Tables -
Analysis 2.9

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 9 Disability.

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 10 Patient Global Impression of Change 'much' or 'very much' improved.
Figures and Tables -
Analysis 2.10

Comparison 2 Lacosamide 400 mg/day versus placebo at end of treatment, Outcome 10 Patient Global Impression of Change 'much' or 'very much' improved.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 1 Pain.
Figures and Tables -
Analysis 3.1

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 1 Pain.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 2 50% pain reduction.
Figures and Tables -
Analysis 3.2

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 2 50% pain reduction.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.
Figures and Tables -
Analysis 3.3

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 3 Sleep problems.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.
Figures and Tables -
Analysis 3.4

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 4 Health‐related quality of life.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 5 Withdrawal due to adverse events.
Figures and Tables -
Analysis 3.5

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 5 Withdrawal due to adverse events.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.
Figures and Tables -
Analysis 3.6

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 6 Dizziness.

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 7 30% pain reduction.
Figures and Tables -
Analysis 3.7

Comparison 3 Levetiracetam up to 3000 mg/day versus placebo at end of treatment, Outcome 7 30% pain reduction.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 1 Pain.
Figures and Tables -
Analysis 4.1

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 1 Pain.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 2 50% pain reduction.
Figures and Tables -
Analysis 4.2

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 2 50% pain reduction.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 3 Fatigue.
Figures and Tables -
Analysis 4.3

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 3 Fatigue.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 4 Sleep problems.
Figures and Tables -
Analysis 4.4

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 4 Sleep problems.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 5 Health‐related quality of life.
Figures and Tables -
Analysis 4.5

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 5 Health‐related quality of life.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 6 Withdrawal due to adverse events.
Figures and Tables -
Analysis 4.6

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 6 Withdrawal due to adverse events.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 7 Serious adverse events.
Figures and Tables -
Analysis 4.7

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 7 Serious adverse events.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 8 Dizziness.
Figures and Tables -
Analysis 4.8

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 8 Dizziness.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 9 30% pain reduction.
Figures and Tables -
Analysis 4.9

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 9 30% pain reduction.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 10 Anxiety.
Figures and Tables -
Analysis 4.10

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 10 Anxiety.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 11 Depression.
Figures and Tables -
Analysis 4.11

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 11 Depression.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 12 Disability.
Figures and Tables -
Analysis 4.12

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 12 Disability.

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 13 Patient Global Impression of Change 'much' or 'very much' improved.
Figures and Tables -
Analysis 4.13

Comparison 4 Pregabalin versus placebo at end of treatment, Outcome 13 Patient Global Impression of Change 'much' or 'very much' improved.

Summary of findings for the main comparison. Pregabalin versus placebo at end of treatment for fibromyalgia

Pregabalin versus placebo at final treatment for fibromyalgia

Patient or population: People with fibromyalgia
Settings: Research centres
Intervention: Pregabalin versus placebo at final treatment

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Anticonvulsants versus placebo at final treatment

50% pain reduction

137 per 1000

217 per 1000
(182 to 260)

RR 1.59
(1.33 to 1.90)

3256
(5 studies)

⊕⊕⊕⊕
high

Absolute risk difference (fewer pain) 8% (95% CI 6% to 11%)

Relative per cent improvement 59% (95% CI 33% to 90%)

NNTB 12 (95% CI 9 to 21)

Patient Global Impression of Change of 'much' or 'very much' improved

279 per 1000

385 per 1000
(344 to 432)

RR 1.38
(1.23 to 1.55)

3183
(5 studies)

⊕⊕⊕⊕
high

Absolute risk difference (more global impression of 'much' and 'very much' improved) 12% (95% CI 4% to 20%)

Relative per cent improvement 38% (95% CI 23% to 55%)

NNTB 9 (95% CI 7 to 15)

Fatigue (1‐50 scale)

Higher scores indicate higher fatigue levels

MAF baseline fatigue score control group

35.6 (standard deviation 8.0) **

The mean fatigue in the intervention groups was
0.17 standard deviations lower
(0.25 to 0.09 lower)

3195
(5 studies)

⊕⊕⊕⊕
high

SMD ‐0.17 (‐0.25 to ‐0.09)

2.7% (95% CI 1.4% to 4.0%) fewer points on the fatigue scale (absolute improvement)

3.8% (95% CI 2.0% to 5.6%) relative improvement

NNTB 13 (95% CI 9 to 25)

Sleep problems (0‐100 scale). Higher scores indicate more sleep problems

MOS baseline overall sleep problem index control group

58.5 (17.8) ***

The mean sleep problems in the intervention groups was
0.35 standard deviations lower
(0.43 to 0.27 lower)

3139
(5 studies)

⊕⊕⊕⊕
high

SMD ‐0.35 (‐0.43 to ‐0.27)

6.2% (95% CI 4.8%

to 7.7%) fewer points on the sleep problem scale (absolute improvement)

10.6% (95% CI 82.% to 13.1%) relative improvement

NNTB 7 (95% CI 5 to 8)

Withdrawal due to adverse events

110 per 1000

185 per 1000
(150 to 229)

RR 1.68
(1.36 to 2.07)

3259
(5 studies)

⊕⊕⊕⊕
high

Absolute risk difference (more withdrawal due to adverse events) 8% (95% CI 5% to 12%)

Relative per cent worsening 68% (95% CI 36% to 107%)

NNTH 13 (95% CI 9 to 23)

Serious adverse events

41 per 1000

42 per 1000
(29 to 61)

RR 1.03
(0.71 to 1.49)

2729
(4 studies)

⊕⊕⊕⊝
moderate 1

Absolute risk difference 0 (95% CI ‐1 to 1)

Relative per cent change 0 (95% CI ‐1 to 1)

Not statistically significant

Dizziness reported to be an adverse event

93 per 1000

350 per 1000
(284 to 429)

RR 3.77
(3.06 to 4.63)

3257
(5 studies)

⊕⊕⊕⊕
high

Absolute risk difference (more dizziness) 28% (95% CI 24% to 32%)

Relative per cent worsening 277% (95% CI 206% to 363%)

NNTH 4 (95% CI 3 to 5)

*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; MAF: Multidimensional Assessment of Fatigue; MOS: Medical Outcomes Study; NNTB: number needed to treat for an additional beneficial outcome; NNTH: number needed to treat for an additional harmful outcome; NRS: numeric rating scale; RR: risk ratio; SMD: standardised mean difference.

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

1 Frequency of serious adverse events not reported by one study.

** Arnold 2008: n = 190 participants; MAF (NRS 1‐50).

*** Arnold 2008: n = 190 participants; MOS Overall Sleep Problems Index (NRS 0‐100).

Figures and Tables -
Summary of findings for the main comparison. Pregabalin versus placebo at end of treatment for fibromyalgia
Comparison 1. Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain Show forest plot

1

119

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

‐0.49 [‐0.86, ‐0.13]

2 50% pain reduction Show forest plot

1

150

Risk Ratio (IV, Random, 95% CI)

1.6 [1.01, 2.53]

3 Sleep problems Show forest plot

1

119

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

‐0.71 [‐1.08, ‐0.34]

4 Health‐related quality of life Show forest plot

1

119

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

‐0.66 [‐1.03, ‐0.29]

5 Withdrawal due to adverse events Show forest plot

1

150

Risk Ratio (IV, Random, 95% CI)

1.71 [0.71, 4.11]

6 Dizziness Show forest plot

1

150

Risk Ratio (IV, Random, 95% CI)

2.71 [1.21, 6.07]

7 30% pain reduction Show forest plot

1

150

Risk Ratio (M‐H, Random, 95% CI)

1.65 [1.10, 2.48]

8 Depression Show forest plot

1

119

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

‐0.52 [‐0.89, ‐0.16]

9 Disability Show forest plot

1

119

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

‐0.94 [‐1.32, ‐0.56]

Figures and Tables -
Comparison 1. Gabapentin flexible 1200 to 2400 mg/day versus placebo at end of treatment
Comparison 2. Lacosamide 400 mg/day versus placebo at end of treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain Show forest plot

1

158

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

‐0.25 [‐0.56, 0.07]

2 Fatigue Show forest plot

1

121

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

‐0.07 [‐0.43, 0.28]

3 Sleep problems Show forest plot

1

158

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

‐0.19 [‐0.51, 0.12]

4 Health‐related quality of life Show forest plot

1

157

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

‐0.15 [‐0.47, 0.16]

5 Serious adverse events Show forest plot

1

159

Risk Ratio (IV, Random, 95% CI)

0.15 [0.01, 2.82]

6 Dizziness Show forest plot

1

159

Risk Ratio (IV, Random, 95% CI)

2.34 [1.08, 5.06]

7 Anxiety Show forest plot

1

133

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

0.0 [‐0.34, 0.34]

8 Depression Show forest plot

1

134

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

0.11 [‐0.23, 0.45]

9 Disability Show forest plot

1

158

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

‐0.19 [‐0.51, 0.12]

10 Patient Global Impression of Change 'much' or 'very much' improved Show forest plot

1

134

Risk Ratio (IV, Random, 95% CI)

1.32 [0.85, 2.04]

Figures and Tables -
Comparison 2. Lacosamide 400 mg/day versus placebo at end of treatment
Comparison 3. Levetiracetam up to 3000 mg/day versus placebo at end of treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain Show forest plot

1

50

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

‐0.21 [‐0.77, 0.36]

2 50% pain reduction Show forest plot

1

66

Risk Ratio (IV, Random, 95% CI)

1.52 [0.43, 5.34]

3 Sleep problems Show forest plot

1

50

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

‐0.16 [‐0.72, 0.40]

4 Health‐related quality of life Show forest plot

1

50

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

0.14 [‐0.42, 0.70]

5 Withdrawal due to adverse events Show forest plot

1

66

Risk Ratio (IV, Random, 95% CI)

0.87 [0.21, 3.56]

6 Dizziness Show forest plot

1

66

Risk Ratio (IV, Random, 95% CI)

1.3 [1.02, 1.66]

7 30% pain reduction Show forest plot

1

66

Risk Ratio (IV, Random, 95% CI)

1.23 [0.65, 2.33]

Figures and Tables -
Comparison 3. Levetiracetam up to 3000 mg/day versus placebo at end of treatment
Comparison 4. Pregabalin versus placebo at end of treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain Show forest plot

5

3252

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

‐0.28 [‐0.35, ‐0.20]

1.1 Pregabalin 150 mg/d

1

175

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

‐0.10 [‐0.45, 0.24]

1.2 Pregabalin 300 mg/d

4

913

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

‐0.24 [‐0.39, ‐0.09]

1.3 Pregabalin 450 mg/d

4

915

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

‐0.36 [‐0.51, ‐0.20]

1.4 Pregabalin 600 mg/d

3

751

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

‐0.30 [‐0.51, ‐0.09]

1.5 Pregabalin flexible 300 or 450 mg/d

1

498

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

‐0.24 [‐0.41, ‐0.06]

2 50% pain reduction Show forest plot

5

3256

Risk Ratio (IV, Random, 95% CI)

1.59 [1.33, 1.90]

2.1 Pregabalin 150 mg/d

1

175

Risk Ratio (IV, Random, 95% CI)

0.92 [0.39, 2.19]

2.2 Pregabalin 300 mg/d

4

915

Risk Ratio (IV, Random, 95% CI)

1.45 [1.03, 2.05]

2.3 Pregabalin 450 mg/d

4

916

Risk Ratio (IV, Random, 95% CI)

1.75 [1.23, 2.49]

2.4 Pregabalin 600 mg/d

3

752

Risk Ratio (IV, Random, 95% CI)

1.51 [1.04, 2.20]

2.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

Risk Ratio (IV, Random, 95% CI)

1.88 [1.26, 2.83]

3 Fatigue Show forest plot

5

3195

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

‐0.17 [‐0.25, ‐0.09]

3.1 Pregabalin 150 mg/d

1

165

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

‐0.20 [‐0.56, 0.15]

3.2 Pregabalin 300 mg/d

4

892

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

‐0.16 [‐0.31, ‐0.01]

3.3 Pregabalin 450 mg/d

4

897

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

‐0.15 [‐0.30, 0.01]

3.4 Pregabalin 600 mg/d

3

743

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

‐0.08 [‐0.25, 0.09]

3.5 Pregabalin flexible 300 or 450 mg/d

1

498

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

‐0.31 [‐0.49, ‐0.14]

4 Sleep problems Show forest plot

5

3193

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

‐0.35 [‐0.43, ‐0.27]

4.1 Pregabalin 150 mg/d

1

163

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

‐0.44 [‐0.80, ‐0.08]

4.2 Pregabalin 300 mg/d

4

897

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

‐0.29 [‐0.44, ‐0.14]

4.3 Pregabalin 450 mg/d

4

893

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

‐0.45 [‐0.63, ‐0.27]

4.4 Pregabalin 600 mg/d

3

744

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

‐0.40 [‐0.57, ‐0.23]

4.5 Pregabalin flexible 300 or 450 mg/d

1

496

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

‐0.20 [‐0.37, ‐0.02]

5 Health‐related quality of life Show forest plot

4

2724

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

‐0.17 [‐0.26, ‐0.09]

5.1 Pregabalin 300 mg/d

3

738

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

‐0.12 [‐0.28, 0.05]

5.2 Pregabalin 450 mg/d

3

737

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

‐0.24 [‐0.41, ‐0.07]

5.3 Pregabalin 600 mg/d

3

751

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

‐0.14 [‐0.31, 0.02]

5.4 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

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

‐0.20 [‐0.37, ‐0.02]

6 Withdrawal due to adverse events Show forest plot

5

3259

Risk Ratio (IV, Random, 95% CI)

1.68 [1.36, 2.07]

6.1 Pregabalin 150 mg/d

1

175

Risk Ratio (IV, Random, 95% CI)

1.19 [0.35, 4.08]

6.2 Pregabalin 300 mg/d

4

917

Risk Ratio (IV, Random, 95% CI)

1.54 [1.02, 2.34]

6.3 Pregabalin 450 mg/d

4

917

Risk Ratio (IV, Random, 95% CI)

2.02 [1.32, 3.09]

6.4 Pregabalin 600 mg/d

3

752

Risk Ratio (IV, Random, 95% CI)

2.53 [1.65, 3.86]

6.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

Risk Ratio (IV, Random, 95% CI)

1.01 [0.65, 1.57]

7 Serious adverse events Show forest plot

4

2729

Risk Ratio (IV, Random, 95% CI)

1.03 [0.71, 1.49]

7.1 Pregabalin 300 mg/d

3

738

Risk Ratio (IV, Random, 95% CI)

0.96 [0.50, 1.86]

7.2 Pregabalin 450 mg/d

3

741

Risk Ratio (IV, Random, 95% CI)

1.03 [0.52, 2.03]

7.3 Pregabalin 600 mg/d

3

752

Risk Ratio (IV, Random, 95% CI)

1.01 [0.55, 1.87]

7.4 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

Risk Ratio (IV, Random, 95% CI)

2.98 [0.31, 28.42]

8 Dizziness Show forest plot

5

3257

Risk Ratio (IV, Random, 95% CI)

3.77 [3.06, 4.63]

8.1 Pregabalin 150 mg/d

1

175

Risk Ratio (IV, Random, 95% CI)

2.44 [0.91, 6.54]

8.2 Pregabalin 300 mg/d

4

916

Risk Ratio (IV, Random, 95% CI)

3.11 [2.09, 4.65]

8.3 Pregabalin 450 mg/d

4

917

Risk Ratio (IV, Random, 95% CI)

3.95 [2.68, 5.82]

8.4 Pregabalin 600 mg/d

3

751

Risk Ratio (IV, Random, 95% CI)

4.00 [2.65, 6.03]

8.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

Risk Ratio (IV, Random, 95% CI)

4.89 [2.89, 8.28]

9 30% pain reduction Show forest plot

5

3259

Risk Ratio (IV, Random, 95% CI)

1.37 [1.22, 1.53]

9.1 Pregabalin 150 mg/d

1

176

Risk Ratio (IV, Random, 95% CI)

1.05 [0.62, 1.77]

9.2 Pregabalin 300 mg/d

4

916

Risk Ratio (IV, Random, 95% CI)

1.35 [1.08, 1.68]

9.3 Pregabalin 450 mg/d

4

917

Risk Ratio (IV, Random, 95% CI)

1.49 [1.20, 1.85]

9.4 Pregabalin 600 mg/d

3

752

Risk Ratio (IV, Random, 95% CI)

1.37 [1.07, 1.76]

9.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

Risk Ratio (IV, Random, 95% CI)

1.32 [1.04, 1.68]

10 Anxiety Show forest plot

5

3214

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

‐0.12 [‐0.20, ‐0.04]

10.1 Pregabalin 150 mg/d

1

166

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

0.07 [‐0.28, 0.42]

10.2 Pregabalin 300 mg/d

4

903

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

‐0.05 [‐0.20, 0.10]

10.3 Pregabalin 450 mg/d

4

900

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

‐0.15 [‐0.30, 0.00]

10.4 Pregabalin 600 mg/d

3

749

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

‐0.16 [‐0.32, 0.01]

10.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

496

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

‐0.17 [‐0.35, 0.01]

11 Depression Show forest plot

5

3212

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

‐0.09 [‐0.16, ‐0.01]

11.1 Pregabalin 150 mg/d

1

165

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

‐0.10 [‐0.45, 0.25]

11.2 Pregabalin 300 mg/d

4

902

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

‐0.05 [‐0.20, 0.10]

11.3 Pregabalin 450 mg/d

4

900

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

‐0.13 [‐0.28, 0.02]

11.4 Pregabalin 600 mg/d

3

749

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

‐0.06 [‐0.23, 0.10]

11.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

496

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

‐0.09 [‐0.27, 0.08]

12 Disability Show forest plot

5

3145

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

‐0.01 [‐0.11, 0.09]

12.1 Pregabalin 150 mg/d

1

166

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

0.11 [‐0.24, 0.47]

12.2 Pregabalin 300 mg/d

4

879

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

0.04 [‐0.11, 0.20]

12.3 Pregabalin 450 mg/d

4

888

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

0.00 [‐0.17, 0.18]

12.4 Pregabalin 600 mg/d

3

714

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

0.03 [‐0.14, 0.21]

12.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

498

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

‐0.29 [‐0.47, ‐0.12]

13 Patient Global Impression of Change 'much' or 'very much' improved Show forest plot

5

3183

Risk Ratio (IV, Random, 95% CI)

1.38 [1.23, 1.55]

13.1 Pregabalin 150 mg/d

1

175

Risk Ratio (IV, Random, 95% CI)

1.30 [0.74, 2.29]

13.2 Pregabalin 300 mg/d

4

886

Risk Ratio (IV, Random, 95% CI)

1.25 [0.99, 1.57]

13.3 Pregabalin 450 mg/d

4

892

Risk Ratio (IV, Random, 95% CI)

1.54 [1.15, 2.06]

13.4 Pregabalin 600 mg/d

3

734

Risk Ratio (IV, Random, 95% CI)

1.37 [1.06, 1.76]

13.5 Pregabalin flexible 300 mg/d or 450 mg/d

1

496

Risk Ratio (IV, Random, 95% CI)

1.44 [1.11, 1.87]

Figures and Tables -
Comparison 4. Pregabalin versus placebo at end of treatment