Scolaris Content Display Scolaris Content Display

Tramadol untuk sakit neuropatik dalam kalangan orang dewasa

Contraer todo Desplegar todo

Referencias

Arbaiza 2007 {published data only}

Arbaiza D, Vidal O. Tramadol in the treatment of neuropathic cancer pain. A double‐blind, placebo‐controlled study. Clinical Drug Investigation 2007;27(1):75‐83. [PUBMED: 17177582]CENTRAL

Boureau 2003 {published data only}

Boureau F, Legallicier P, Kabir‐Ahmadi M. Tramadol in post‐herpetic neuralgia: a randomized, double‐blind, placebo‐controlled trial. Pain 2003;104(1‐2):323‐31. [PUBMED: 12855342]CENTRAL

Harati 1998 {published data only}

Harati Y, Gooch C, Swenson M, Edelman S, Greene D, Raskin P, et al. Double‐blind randomized trial of tramadol for the treatment of the pain of diabetic neuropathy. Neurology 1998;50(6):1842‐6. [PUBMED: 9633738]CENTRAL

Norrbrink 2009 {published data only}

Norrbrink C, Lundeberg T. Tramadol in neuropathic pain after spinal cord injury: a randomized, double‐blind, placebo‐controlled trial. Clinical Journal of Pain 2009;25(3):177‐84. [DOI: 10.1097/AJP.0b013e31818a744d; PUBMED: 19333166]CENTRAL

Sindrup 1999 {published data only}

Sindrup SH, Andersen G, Madsen C, Smith T, Brøsen K, Jensen TS. Tramadol relieves pain and allodynia in polyneuropathy: a randomised, double‐blind, controlled trial. Pain 1999;83(1):85‐90. [DOI: 10.1016/S0304‐3959(99)00079‐2; PUBMED: 10506675]CENTRAL
Sindrup SH, Madsen C, Brøsen K, Jensen TS. The effect of tramadol in painful polyneuropathy in relation to serum drug and metabolite levels. Clinical Pharmacology and Therapeutics 1999;66(6):636‐41. [DOI: 10.1053/cp.1999.v66.103171001; PUBMED: 10613620]CENTRAL

Sindrup 2012 {published data only}

Sindrup SH, Konder R, Lehmann R, Meier T, Winkel M, Ashworth J, et al. Randomized controlled trial of the combined monoaminergic and opioid investigational compound GRT9906 in painful polyneuropathy. European Journal of Pain 2012;16(6):849‐59. [DOI: 10.1002/j.1532‐2149.2011.00069.x; PUBMED: 22337471]CENTRAL

Ashry 2001 {published data only}

Ashry H. Double‐blind randomized trial of tramadol for the treatment of the pain of diabetic neuropathy. Foot and Ankle Quarterly ‐ The Seminar Journal 2001;14(4):129‐31. CENTRAL

Attal 2001 {published data only}

Attal N. Pharmacologic treatment of neuropathic pain. Acta Neurologica Belgica 2001;101(1):53‐64. CENTRAL

Benedetti 1998 {published data only}

Benedetti F, Vighetti S, Amanzio M, Casadio C, Oliaro A, Bergamasco B, et al. Dose‐response relationship of opioids in nociceptive and neuropathic postoperative pain. Pain 1998;74(2‐3):205‐11. [MEDLINE: 1998057182]CENTRAL

Erdine 1997 {published data only}

Erdine S, Yucel A, Ozyalcin S. Efficacy of tramadol hydrochloride in chronic painful diabetic neuropathy: a double‐blind placebo controlled study. Proceedings of the 8th World Congress on Pain. Seattle: IASP Press, 1997:371. CENTRAL

Gobel 1995 {published data only}

Gobel H, Stadler TH. Treatment of pain due to postherpetic neuralgia with tramadol. Clinical Drug Investigation 1995;10(4):208‐14. CENTRAL

Harati 1999 {published data only}

Harati Y. Tramadol for the treatment of the pain of diabetic neuropathy [Correspondence]. Neurology 1999;52(6):1300. CENTRAL

Harati 2000 {published data only}

Harati Y, Gooch C, Swenson M, Edelman SV, Greene D, Raskin P, et al. Maintenance of long‐term effectiveness of tramadol in treatment of the pain of diabetic neuropathy. Journal of Diabetes and its Complications 2000;14(2):65‐70. [PUBMED: 10959067]CENTRAL

Herrera Silva 2001 {published data only}

Herrera Silva J. The use of oral opioids in neuropathic pain: development of new tramadol and morphine formulations [Utilidad de los opioides orales en dolor neuropático. Apariciónde nuevas formulaciones de tramadoly morfina]. Revista de la Sociedad Espanola del Dolor 2001;8(Suppl):32‐4. CENTRAL

Leppert 2001 {published data only}

Leppert W. Analgesic efficacy and side effects of oral tramadol and morphine administered orally in the treatment of cancer pain. Nowotwory 2001;51(3):257‐66. CENTRAL

Moulin 1999 {published data only}

Moulin D. Tramadol for the treatment of the pain of diabetic neuropathy [Correspondence]. Neurology 1999;52(6):1301. CENTRAL

NCT00610155 {published data only}

NCT00610155. A methodology study of brain imaging of pain‐killers in post‐traumatic neuropathic pain patients [A methodology study to assess the feasibility of using functional magnetic resonance imaging (fMRI) to quantify the effects of analgesic drugs in post‐traumatic neuropathic pain subjects]. clinicaltrials.gov/ct2/show/NCT00610155 (first received 14 January 2008). [CTG: NCT00610155]CENTRAL

Saxena 2013 {published data only}

Nasare NV, Banerjee BD, Deshmukh PS, Mediratta PK, Ahmed RS, Saxena AK, et al. Neuropathic pain symptoms of post herpetic neuralgia patients with CYP2D6 polymorphism undergoing tramadol treatment. International Journal of Pharmaceutical Sciences and Research 2015;6(4):1489‐501. [DOI: 10.13040/IJPSR.0975‐8232.6(4).1489‐01]CENTRAL
Saxena AK, Nasare N, Jain S, Dhakate G, Ahmed RS, Bhattacharya SN, et al. A randomized, prospective study of efficacy and safety of oral tramadol in the management of post‐herpetic neuralgia in patients from north India. Pain Practice 2013;13(4):264‐75. [DOI: 10.1111/j.1533‐2500.2012.00583.x]CENTRAL

Xiao 2004 {published data only}

Xiao LZ, Zhang DR, Jiang J, Zhang KL, Zhang M, Zhu HQ. Feasibility of transdermal fentanyl for pain relief of herpes zoster and postherpetic neuralgia. Zhongguo Linchuang Kangfu 2004;8(2):216‐7. CENTRAL

References to studies awaiting assessment

Ho 2009 {published data only}

Ho TW, Backonja M, Ma J, Leibensperger H, Froman S, Polydefkis M. Efficient assessment of neuropathic pain drugs in patients with small fiber sensory neuropathies. Pain 2009;141(1‐2):19‐24. [DOI: 10.1016/j.pain.2008.07.013]CENTRAL

AlBalawi 2013

AlBalawi Z, McAlister FA, Thorlund K, Wong M, Wetterslev J. Random error in cardiovascular meta‐analyses: how common are false positive and false negative results?. International Journal of Cardiology 2013;168(2):1102‐7. [DOI: 10.1016/j.ijcard.2012.11.048]

Baron 2012

Baron R, Wasner G, Binder A. Chronic pain: genes, plasticity, and phenotypes. Lancet Neurology 2012;11(1):19‐21. [DOI: 10.1016/S1474‐4422(11)70281‐]

Beakley 2015

Beakley BD, Kaye AM, Kaye AD. Tramadol, pharmacology, side effects, and serotonin syndrome: a review. Pain Physician 2015;18(4):395‐400. [PUBMED: 26218943]

Berger 2004

Berger A, Dukes EM, Oster G. Clinical characteristics and economic costs of patients with painful neuropathic disorders. Journal of Pain 2004;5(3):143‐9. [DOI: 10.1016/j.jpain.2003.12.004]

Berger 2009

Berger A, Toelle T, Sadosky A, Dukes E, Edelsberg J, Oster G. Clinical and economic characteristics of patients with painful neuropathic disorders in Germany. Pain Practice 2009;9(1):8‐17. [DOI: 10.1111/j.1533‐2500.2008.00244.x]

Berger 2012

Berger A, Sadosky A, Dukes E, Edelsberg J, Oster G. Clinical characteristics and patterns of healthcare utilization in patients with painful neuropathic disorders in UK general practice: a retrospective cohort study. BMC Neurology 2012;12:8. [DOI: 10.1186/1471‐2377‐12‐8]

Bouhassira 2008

Bouhassira D, Lantéri‐Minet M, Attal N, Laurent B, Touboul C. Prevalence of chronic pain with neuropathic characteristics in the general population. Pain 2008;136(3):380‐7. [DOI: 10.1016/j.pain.2007.08.013]

Bozkurt 2005

Bozkurt P. Use of tramadol in children. Paediatric Anaesthesia 2005;15(12):1041‐7. [DOI: 10.1111/j.1460‐9592.2005.01738.x]

Brok 2009

Brok J, Thorlund K, Wetterslev J, Gluud C. Apparently conclusive meta‐analyses may be inconclusive‐‐Trial sequential analysis adjustment of random error risk due to repetitive testing of accumulating data in apparently conclusive neonatal meta‐analyses. International Journal of Epidemiology 2009;38(1):287‐98. [DOI: 10.1093/ije/dyn188]

Bush 2015

Bush DM. Emergency department visits for drug misuse or abuse involving the pain medication tramadol. The CBHSQ Report. Rockville (MD): Substance Abuse and Mental Health Services Administration (US); 2013‐.. The CBHSQ, 2015; Vol. May 14.

CADTH 2015

Tramadol for the management of pain in adult patients: a review of the clinical effectiveness. Canadian Agency for Drugs and Technologies in Health 2 February 2015; Vol. www.cadth.ca/sites/default/files/pdf/htis/may‐2015/RC0627‐Tramadol‐pain‐Final.pdf (accessed 9 January 2017).

Calvo 2012

Calvo M, Dawes JM, Bennett DL. The role of the immune system in the generation of neuropathic pain. Lancet Neurology 2012;11(7):629‐42. [DOI: 10.1016/S1474‐4422(12)70134‐5]

Cepeda 2006

Cepeda MS, Camargo F, Zea C, Velencia L. Tramadol for osteoarthritis. Cochrane Database of Systematic Reviews 2006, Issue 3. [DOI: 10.1002/14651858.CD005522.pub2]

Chaparro 2012

Chaparro LE, Wiffen PJ, Moore RA, Gilron I. Combination pharmacotherapy for the treatment of neuropathic pain in adults. Cochrane Database of Systematic Reviews 2012, Issue 7. [DOI: 10.1002/14651858.CD008943.pub2]

Dechartres 2013

Dechartres A, Trinquart L, Boutron I, Ravaud P. Influence of trial sample size on treatment effect estimates: meta‐epidemiological study. BMJ 2013;346:f2304. [DOI: 10.1136/bmj.f2304]

Dechartres 2014

Dechartres A, Altman DG, Trinquart L, Boutron I, Ravaud P. Association between analytic strategy and estimates of treatment outcomes in meta‐analyses. JAMA 2014;312(6):623‐30. [DOI: 10.1001/jama.2014.8166]

Demant 2014

Demant DT, Lund K, Vollert J, Maier C, Segerdahl M, Finnerup NB, et al. The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double‐blind, placebo‐controlled phenotype‐stratified study. Pain 2014;155(11):2263‐73. [DOI: 10.1016/j.pain.2014.08.014]

Derry 2012

Derry S, Moore RA. Topical capsaicin (low concentration) for chronic neuropathic pain in adults. Cochrane Database of Systematic Reviews 2012, Issue 9. [DOI: 10.1002/14651858.CD010111]

Derry 2013

Derry S, Sven‐Rice A, Cole P, Tan T, Moore RA. Topical capsaicin (high concentration) for chronic neuropathic pain in adults. Cochrane Database of Systematic Reviews 2013, Issue 2. [DOI: 10.1002/14651858.CD007393.pub3]

Derry 2014

Derry S, Wiffen PJ, Moore RA, Quinlan J. Topical lidocaine for neuropathic pain in adults. Cochrane Database of Systematic Reviews 2014, Issue 7. [DOI: 10.1002/14651858.CD010958.pub2]

Dworkin 2008

Dworkin RH, Turk DC, Wyrwich KW, Beaton D, Cleeland CS, Farrar JT, et al. Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations. Journal of Pain 2008;9(2):105‐21. [DOI: 10.1016/j.jpain.2007.09.005]

Dworkin 2013

Dworkin RH, O'Connor AB, Kent J, Mackey SC, Raja SN, Stacey BR, et al. Interventional management of neuropathic pain: NeuPSIG recommendations. Pain 2013;154(11):2249‐61. [DOI: 10.1016/j.pain.2013.06.004]

Edwards 2002

Edwards JE, McQuay HJ, Moore RA. Combination analgesic efficacy: individual patient data meta‐analysis of single‐dose oral tramadol plus acetaminophen in acute postoperative pain. Journal of Pain and Symptom Management 2002;23(2):121‐30. [DOI: 10.1016/S0885‐3924(01)00404‐3]

EPOC 2015

Effective Practice, Organisation of Care (EPOC). 23. Worksheets for preparing a Summary of Findings using GRADE. EPOC Resources for review authors. Oslo: Norwegian Knowledge Centre for the Health Services; 2015. Available at: http://epoc.cochrane.org/epoc‐specific‐resources‐review‐authors (accessed 9 January 2017).

Finnerup 2013

Finnerup NB, Scholz J, Attal N, Baron R, Haanpää M, Hansson P, et al. Neuropathic pain needs systematic classification. European Journal of Pain 2013;17(7):953‐6. [DOI: 10.1002/j.1532‐2149.2012.00282.x]

Finnerup 2015

Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, Dworkin RH, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta‐analysis. Lancet Neurology 2015;14(2):162‐73. [DOI: 10.1016/S1474‐4422(14)70251‐0]

Gan 2007

Gan SH, Ismail R, Wan Adnan WA, Zulmi W. Impact of CYP2D6 genetic polymorphism on tramadol pharmacokinetics and pharmacodynamics. Molecular Diagnosis and Therapy 2007;11(3):171‐81. [DOI: 10.1007/BF03256239]

Gaskell 2016

Gaskell H, Derry S, Stannard C, Moore RA. Oxycodone for neuropathic pain in adults. Cochrane Database of Systematic Reviews 2016, Issue 7. [DOI: 10.1002/14651858.CD010692.pub3]

Grond 2004

Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clinical Pharmacokinetics 2004;43(13):879‐932. [DOI: 10.2165/00003088‐200443130‐00004]

Gustorff 2008

Gustorff B, Dorner T, Likar R, Grisold W, Lawrence K, Schwarz F, et al. Prevalence of self‐reported neuropathic pain and impact on quality of life: a prospective representative survey. Acta Anaesthesiologica Scandinavica 2008;52(1):132‐6. [DOI: 10.1111/j.1399‐6576.2007.01486.x]

Guyatt 2013a

Guyatt G, Oxman AD, Sultan S, Brozek J, Glasziou P, Alonso‐Coello P, et al. GRADE guidelines: 11. Making an overall rating of confidence in effect estimates for a single outcome and for all outcomes. Journal of Clinical Epidemiology 2013;66(2):151‐7. [DOI: 10.1016/j.jclinepi.2012.01.006]

Guyatt 2013b

Guyatt GH, Oxman AD, Santesso N, Helfand M, Vist G, Kunz R, et al. GRADE guidelines: 12. Preparing summary of findings tables‐binary outcomes. Journal of Clinical Epidemiology 2013;66(2):158‐72. [DOI: 10.1016/j.jclinepi.2012.01.012]

Hall 2008

Hall GC, Carroll D, McQuay HJ. Primary care incidence and treatment of four neuropathic pain conditions: a descriptive study, 2002‐2005. BMC Family Practice 2008;9:26. [DOI: 10.1186/1471‐2296‐9‐26]

Helfert 2015

Helfert SM, Reimer M, Höper J, Baron R. Individualized pharmacological treatment of neuropathic pain. Clinical Pharmacology and Therapeutics 2015;97(2):135‐42. [DOI: 10.1002/cpt.19]

Higgins 2003

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

Higgins 2011

Higgins JPT, Altman DG, Sterne JAC (editors). Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Hoffman 2010

Hoffman DL, Sadosky A, Dukes EM, Alvir J. How do changes in pain severity levels correspond to changes in health status and function in patients with painful diabetic peripheral neuropathy?. Pain 2010;149(2):194‐201. [DOI: 10.1016/j.pain.2009.09.017]

Jadad 1996

Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary?. Controlled Clinical Trials 1996;17(1):1‐12. [DOI: 10.1016/0197‐2456(95)00134‐4]

Jensen 2011

Jensen TS, Baron R, Haanpää M, Kalso E, Loeser JD, Rice AS, et al. A new definition of neuropathic pain. Pain2011; Vol. 152, issue 10:2204‐5. [DOI: 10.1016/j.pain.2011.06.017]

Kalso 2013

Kalso E, Aldington DJ, Moore RA. Drugs for neuropathic pain. BMJ 2013;347:f7339. [DOI: 10.1136/bmj.f7339]

Katusic 1991

Katusic S, Williams DB, Beard CM, Bergstralh EJ, Kurland LT. Epidemiology and clinical features of idiopathic trigeminal neuralgia and glossopharyngeal neuralgia: similarities and differences, Rochester, Minnesota 1945‐1984. Neuroepidemiology 1991;10(5‐6):276‐81. [DOI: 10.1159/000110284]

Koopman 2009

Koopman JS, Dieleman JP, Huygen FJ, de Mos M, Martin CG, Sturkenboom MC. Incidence of facial pain in the general population. Pain 2009;147(1‐3):122‐7. [DOI: 10.1016/j.pain.2009.08.023]

L'Abbé 1987

L'Abbé KA, Detsky AS, O'Rourke K. Meta‐analysis in clinical research. Annals of Internal Medicine 1987;107(2):224‐33. [DOI: 10.7326/0003‐4819‐107‐2‐224]

Lintz 1998

Lintz W, Barth H, Osterloh G, Schmidt‐Bothelt E. Pharmacokinetics of tramadol and bioavailability of enteral tramadol formulations. 3rd Communication: suppositories. Arzneimittel‐Forschung 1998;48(9):889‐99. [PUBMED: 9793614]

Lunn 2014

Lunn MP, Hughes RA, Wiffen PJ. Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia. Cochrane Database of Systematic Reviews 2014, Issue 1. [DOI: 10.1002/14651858.CD007115.pub3]

Martindale 2016

Martindale: the complete drug reference. Tramadol hydrochloride. www.medicinescomplete.com/mc/ (accessed 9 January 2017). 38th. Pharmaceutical Press, 2016.

McQuay 1996

McQuay H, Carroll D, Moore A. Variation in the placebo effect in randomised controlled trials of analgesics: all is as blind as it seems. Pain 1996;64(2):331‐5. [DOI: 10.1016/0304‐3959(95)00116‐6]

McQuay 1998

McQuay HJ, Moore RA. An Evidence‐Based Resource for Pain Relief. Oxford: Oxford University Press, 1998. [ISBN: 0‐19‐263048‐2]

McQuay 2007

McQuay HJ, Smith LA, Moore RA. Chronic pain. In: Stevens A, Raftery J, Mant J, Simpson S editor(s). Health Care Needs Assessment, 3rd Series. Oxford: Radcliffe Publishing, 2007. [ISBN: 978‐1‐84619‐063‐6]

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group (2009). Preferred reporting items for systematic reviews and meta‐analyses: The PRISMA Statement. PLoS Medicine 2009;6(7):e1000097. [DOI: 10.1371/journal.pmed1000097]

Moore 1997

Moore RA, McQuay HJ. Single‐patient data meta‐analysis of 3453 postoperative patients: oral tramadol versus placebo, codeine and combination analgesics. Pain 1997;69(3):287‐94. [DOI: 10.1016/S0304‐3959(96)03291‐5]

Moore 1998

Moore RA, Gavaghan D, Tramèr MR, Collins SL, McQuay HJ. Size is everything ‐ large amounts of information are needed to overcome random effects in estimating direction and magnitude of treatment effects. Pain 1998;78(3):209‐16. [DOI: 10.1016/S0304‐3959(98)00140‐7]

Moore 2008

Moore RA, Barden J, Derry S, McQuay HJ. Managing potential publication bias. In: McQuay HJ, Kalso E, Moore RA editor(s). Systematic Reviews in Pain Research: Methodology Refined. Seattle: IASP Press, 2008:15‐24. [ISBN: 978‐0‐931092‐69‐5]

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 RA, Eccleston C, Derry S, Wiffen P, Bell RF, Straube S, et al. "Evidence" in chronic pain ‐ establishing best practice in the reporting of systematic reviews. Pain 2010;150(3):386‐9. [DOI: 10.1016/j.pain.2010.05.011]

Moore 2010b

Moore RA, Straube S, Paine J, Phillips CJ, Derry S, McQuay HJ. Fibromyalgia: moderate and substantial pain intensity reduction predicts improvement in other outcomes and substantial quality of life gain. Pain 2010;149(2):360‐4. [DOI: 10.1016/j.pain.2010.02.039]

Moore 2010c

Moore RA, Smugar SS, Wang H, Peloso PM, Gammaitoni A. Numbers‐needed‐to‐treat analyses ‐ do timing, dropouts, and outcome matter? Pooled analysis of two randomized, placebo‐controlled chronic low back pain trials. Pain 2010;151(3):592‐7. [DOI: 10.1016/j.pain.2010.07.013]

Moore 2010d

Moore RA, Moore OA, Derry S, Peloso PM, Gammaitoni AR, Wang H. Responder analysis for pain relief and numbers needed to treat in a meta‐analysis of etoricoxib osteoarthritis trials: bridging a gap between clinical trials and clinical practice. Annals of the Rheumatic Diseases 2010;69(2):374‐9. [DOI: 10.1136/ard.2009.107805]

Moore 2011a

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 2011b

Moore RA, Straube S, Paine J, Derry S, McQuay HJ. Minimum efficacy criteria for comparisons between treatments using individual patient meta‐analysis of acute pain trials: examples of etoricoxib, paracetamol, ibuprofen, and ibuprofen/paracetamol combinations after third molar extraction. Pain 2011;152(5):982‐9. [DOI: 10.1016/j.pain.2010.11.030]

Moore 2012

Moore RA, Straube S, Eccleston C, Derry S, Aldington D, Wiffen P, et al. Estimate at your peril: imputation methods for patient withdrawal can bias efficacy outcomes in chronic pain trials using responder analyses. Pain 2012;153(2):265‐8. [DOI: 10.1016/j.pain.2011.10.004]

Moore 2013a

Moore RA, Straube S, Aldington D. Pain measures and cut‐offs ‐ 'no worse than mild pain' as a simple, universal outcome. Anaesthesia 2013;68(4):400‐12. [DOI: 10.1111/anae.12148]

Moore 2013b

Moore A, Derry S, Eccleston C, Kalso E. Expect analgesic failure; pursue analgesic success. BMJ 2013;346:f2690. [DOI: 10.1136/bmj.f2690]

Moore 2014a

Moore RA, Derry S, Taylor RS, Straube S, Phillips CJ. The costs and consequences of adequately managed chronic non‐cancer pain and chronic neuropathic pain. Pain Practice 2014;14(1):79‐94. [DOI: 10.1111/papr.12050]

Moore 2014b

Moore RA, Cai N, Skljarevski V, Tölle TR. Duloxetine use in chronic painful conditions ‐ individual patient data responder analysis. European Journal of Pain 2014;18(1):67‐75. [DOI: 10.1002/j.1532‐2149.2013.00341.x]

Moore 2014c

Moore RA, Wiffen PJ, Derry S, Toelle T, Rice AS. Gabapentin for chronic neuropathic pain and fibromyalgia in adults. Cochrane Database of Systematic Reviews 2014, Issue 4. [DOI: 10.1002/14651858.CD007938.pub3]

Moore 2015a

Moore RA, Chi CC, Wiffen PJ, Derry S, Rice ASC. Oral nonsteroidal anti‐inflammatory drugs for neuropathic pain. Cochrane Database of Systematic Reviews 2015, Issue 10. [DOI: 10.1002/14651858.CD010902.pub2]

Moore 2015b

Moore RA, Derry S, Aldington D, Cole P, Wiffen PJ. Amitriptyline for neuropathic pain in adults. Cochrane Database of Systematic Reviews 2015, Issue 7. [DOI: 10.1002/14651858.CD008242.pub3]

Moore 2015c

Moore RA, Wiffen PJ, Eccleston C, Derry S, Baron R, Bell RF, et al. Systematic review of enriched enrolment, randomised withdrawal trial designs in chronic pain: a new framework for design and reporting. Pain 2015;156(8):1382‐95. [DOI: 10.1097/j.pain.0000000000000088]

NICE 2013

National Institute for Health and Care Excellence (NICE). Neuropathic pain ‐ pharmacological management: the pharmacological management of neuropathic pain in adults in non‐specialist settings, 2013. www.nice.org.uk/guidance/cg173 (accessed 9 January 2017).

Nüesch 2010

Nüesch E, Trelle S, Reichenbach S, Rutjes AW, Tschannen B, Altman DG, et al. Small study effects in meta‐analyses of osteoarthritis trials: meta‐epidemiological study. BMJ 2010;341:c3515. [DOI: 10.1136/bmj.c3515]

O'Brien 2010

O'Brien EM, Staud RM, Hassinger AD, McCulloch RC, Craggs JG, Atchison JW, et al. Patient‐centered perspective on treatment outcomes in chronic pain. Pain Medicine 2010;11(1):6‐15. [DOI: 10.1111/j.1526‐4637.2009.00685.]

PaPaS 2012

Cochrane Pain, Palliative and Supportive Care Group (PaPaS) author and referee guidance. papas.cochrane.org/papas‐documents (accessed 9 January 2017).

Radbruch 2013

Radbruch L, Glaeske G, Grond S, Münchberg F, Scherbaum N, Storz E, et al. Topical review on the abuse and misuse potential of tramadol and tilidine in Germany. Substance Abuse 2013;34(3):313‐20. [DOI: 10.1080/08897077.2012.735216]

Rappaport 1994

Rappaport ZH, Devor M. Trigeminal neuralgia: the role of self‐sustaining discharge in the trigeminal ganglion. Pain 1994;56(2):127‐38. [DOI: 10.1016/0304‐3959(94)90086‐8]

Reeves 2008

Reeves RR, Burke RS. Tramadol: basic pharmacology and emerging concepts. Drugs of Today (Barcelona, Spain: 1998) 2008;44(11):827‐36. [DOI: 10.1358/dot.2008.44.11.1289441]

RevMan 2014 [Computer program]

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

Roberts 2015

Roberts I, Ker K, Edwards P, Beecher D, Manno D, Sydenham E. The knowledge system underpinning healthcare is not fit for purpose and must change. BMJ 2015;350:h2463. [DOI: 10.1136/bmj.h2463]

Schünemann 2011a

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

Schünemann 2011b

Schünemann HJ, Oxman AD, Higgins JPT, Vist GE, Glasziou P, Guyatt GH. Chapter 11: Presenting results and ‘Summary of findings' tables. In: Higgins JPT, Green S (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Scott 2000

Scott LJ, Perry CM. Tramadol: a review of its use in perioperative pain. Drugs 2000;60(1):139‐76. [DOI: 10.2165/00003495‐200060010‐00008]

Scott 2006

Scott FT, Johnson RW, Leedham‐Green M, Davies E, Edmunds WJ, Breuer J. The burden of herpes zoster: a prospective population based study. Vaccine 2006;24(9):1308‐14. [DOI: 10.1016/j.vaccine.2005.09.026]

Stannard 2016

Stannard C, Gaskell H, Derry S, Aldington D, Cole P, Cooper TE, et al. Hydromorphone for neuropathic pain in adults. Cochrane Database of Systematic Reviews 2016, Issue 5. [DOI: 10.1002/14651858.CD011604.pub2]

Straube 2008

Straube S, Derry S, McQuay HJ, Moore RA. Enriched enrolment: definition and effects of enrichment and dose in trials of pregabalin and gabapentin in neuropathic pain. A systematic review. British Journal of Clinical Pharmacology 2008;66(2):266‐75. [DOI: 10.1111/j.1365‐2125.2008.03200.]

Straube 2010

Straube S, Derry S, Moore RA, Paine J, McQuay HJ. Pregabalin in fibromyalgia ‐ responder analysis from individual patient data. BMC Musculoskeletal Disorders 2010;11:150. [DOI: 10.1186/1471‐2474‐11‐150]

Sultan 2008

Sultan A, Gaskell H, Derry S, Moore RA. Duloxetine for painful diabetic neuropathy and fibromyalgia pain: systematic review of randomised trials. BMC Neurology 2008;8:29. [DOI: 10.1186/1471‐2377‐8‐29]

Tassinari 2011

Tassinari D, Drudi F, Rosati M, Tombesi P, Sartori S, Maltoni M. The second step of the analgesic ladder and oral tramadol in the treatment of mild to moderate cancer pain: a systematic review. Palliative Medicine 2011;25(5):410‐23. [DOI: 10.1177/0269216311405090]

Thorlund 2011

Thorlund K, Imberger G, Walsh M, Chu R, Gluud C, Wetterslev J, et al. The number of patients and events required to limit the risk of overestimation of intervention effects in meta‐analysis‐‐a simulation study. PLoS One 2011;6(10):e25491. [DOI: 10.1371/journal.pone.0025491]

Torrance 2006

Torrance N, Smith BH, Bennett MI, Lee AJ. The epidemiology of chronic pain of predominantly neuropathic origin. Results from a general population survey. Journal of Pain 2006;7(4):281‐9. [DOI: 10.1016/j.jpain.2005.11.008]

Treede 2008

Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, et al. Neuropathic pain: redefinition and a grading system for clinical and research purposes. Neurology 2008;70(18):1630‐5. [DOI: 10.1212/01.wnl.0000282763.29778.59]

Turner 2013

Turner RM, Bird SM, Higgins JP. The impact of study size on meta‐analyses: examination of underpowered studies in Cochrane reviews. PLoS One 2013;8(3):e59202. [DOI: 10.1371/journal.pone.0059202]

Van Hecke 2014

van Hecke O, Austin SK, Khan RA, Smith BH, Torrance N. Neuropathic pain in the general population: a systematic review of epidemiological studies. Pain 2014;155(4):654‐62. [DOI: 10.1016/j.pain.2013.11.013]

Van Hoek 2009

van Hoek AJ, Gay N, Melegaro A, Opstelten W, Edmunds WJ. Estimating the cost‐effectiveness of vaccination against herpes zoster in England and Wales. Vaccine 2009;27(9):1454‐67. [DOI: 10.1016/j.vaccine.2008.12.024]

von Hehn 2012

von Hehn CA, Baron R, Woolf CJ. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron 2012;73(4):638‐52. [DOI: 10.1016/j.neuron.2012.02.008]

Vos 2012

Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati M, et al. Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990‐2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012;380(9859):2163‐96. [DOI: 10.1016/S0140‐6736(12)61729‐2]

WHO 2016

World Health Organization (WHO). WHO analgesic ladder. www.who.int/cancer/palliative/painladder/en/ (accessed 9 January 2017).

Wiffen 2013

Wiffen PJ, Derry S, Moore RA, Aldington D, Cole P, Rice ASC, et al. Antiepileptic drugs for neuropathic pain and fibromyalgia ‐ an overview of Cochrane reviews. Cochrane Database of Systematic Reviews 2013, Issue 11. [DOI: 10.1002/14651858.CD010567.pub2]

References to other published versions of this review

Dühmke 2002

Dühmke R, Hollingshead J, Cornblath D. Tramadol for neuropathic pain. Cochrane Database of Systematic Reviews 2002, Issue 2. [DOI: 10.1002/14651858.CD003726]

Dühmke 2004

Dühmke RM, Cornblath DD, Hollingshead JRF. Tramadol for neuropathic pain. Cochrane Database of Systematic Reviews 2004, Issue 2. [DOI: 10.1002/14651858.CD003726.pub2]

Hollingshead 2006

Hollingshead J, Dühmke RM, Cornblath DR. Tramadol for neuropathic pain. Cochrane Database of Systematic Reviews 2006, Issue 3. [DOI: 10.1002/14651858.CD003726.pub3]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Arbaiza 2007

Methods

Randomised ("matched pair"), double‐blind, parallel‐group, placebo‐controlled

Duration: 45 days

Assessed at baseline, 15, 30, 45 days

Participants

Cancer‐related or cancer treatment‐related neuropathic pain of ≥ moderate intensity for ≥ 3 months, aged 18‐60 years

Exclusion: pain mainly somatic, visceral or sympathetically maintained; scheduled for surgery, radiotherapy, chemotherapy, hormone therapy; use of tricyclic antidepressants, tramadol or any opioid; respiratory failure, chronic obstructive pulmonary disease, intracranial hypertension; Hx psychiatric illness or dependency on alcohol or drugs

N = 36
M 14, F 22
Mean age 50 years
Mean baseline PI: 7/10

Interventions

Tramadol 1 mg/kg bodyweight every 6 h; increased to 1.5 mg/kg every 6 h if relief inadequate, n = 18
Placebo, n = 18

Participants could continue with previous antiepileptic analgesic therapy ‐ and could reduce dose during study

Rescue medication: paracetamol 500 mg/d

Outcomes

PI: 0‐10 NRS
Reduction in use of antiepileptics: 0 = no need for antiepileptics, 5 = 100% analgesic use at first assessment
Adverse events

Notes

Peru. Sponsor: Grunenthal Laboratories, Peru

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants with similar pain syndromes were paired, then "randomly assigned using a computer program"

Allocation concealment (selection bias)

Unclear risk

Method not described, but effectively, the first of pair was randomised, leaving a possibility of unconcealed allocation

Blinding (performance bias and detection bias)
All outcomes

Low risk

Treatments supplied in identical 10 ml bottles, "distinguished only by labels"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation not mentioned, approximately 30% withdrawals, with different reasons between groups

Size

High risk

< 50 participants per treatment arm (18)

Boureau 2003

Methods

Multicentre, randomised, double‐blind, parallel‐group, placebo‐controlled

Duration 6 weeks
Assessments at 1, 8, 15, 22, 43 days

Participants

Postherpetic neuralgia ≥ 3 months and ≤ 1 year, PI ≥ 40/100, aged 18‐85 years
Exclusion: seizures; cerebral tumour or recent cranial trauma; severe hepatic, renal, cardiac, respiratory pathology; contraindication to tramadol or opioids; Hx depression, drug abuse

N = 127 (125 in ITT population, 108 in PP)
M 35, F 92
Mean age ˜67 years (35‐85)

Mean baseline PI: 60/100

Interventions

Tramadol SR 100 mg taken in evening, n = 64
Placebo, n = 63
Dose could be increased to maximum 400 mg (≤ 75 years) or 300 mg (75+ years) taken as divided dose in morning and evening

Rescue medication: paracetamol to maximum 3 g/d

No MAO within 15 days, or antidepressants, anticonvulsants, opioid analgesics or local/general anaesthetics within 7 days

Outcomes

PI in last 24 h (daily): 100 mm VAS, 5‐point VRS
PGE: % reduction from baseline
Use of rescue medication
Adverse events

Notes

France. Sponsor: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated 4‐block centralised randomisation list

Allocation concealment (selection bias)

Unclear risk

Method not described

Blinding (performance bias and detection bias)
All outcomes

Low risk

"treatments were identical with regard to appearance"

Incomplete outcome data (attrition bias)
All outcomes

High risk

Missing data on VAS and VRS over the 6th week were replaced by data available from the last 7 observations before the final visit (or the visit before premature discontinuation), not including more than 13 days before the end visit. Essentially an LOCF analysis

Size

Unclear risk

50‐199 participants per treatment arm (63, 64)

Harati 1998

Methods

Multicenter, randomised, double‐blind, placebo‐controlled, parallel‐group
Duration: 6 weeks
Assessed at 1, 14, 28, 42 days

Participants

Peripheral diabetic neuropathy (HbA1 < 14%), distal, symmetric, > 3 months, PI moderate without analgesics, aged 18 years and over
Exclusion: contraindication or previous use of tramadol; cause other than diabetes; other pain > neuropathic pain; clinically significant medical conditions; use of multiple daily doses of opioids or regular mexiletine; amputations; open ulcers; Hx drug or alcohol abuse

N = 131 (127 for efficacy)
M 78, F 53
Mean age 57 years (32‐85)
Baseline pain 2.5 (scale 0‐4)

Interventions

Tramadol starting at 50 mg/d, increasing to 200 mg/d on day 10, then increased again as required from day 14 to maximum 400 mg/d by day 28, then stable; minimum 100 mg/d from day 14 to end of study, n = 65
Placebo, n = 66

Divided doses, given 4 x daily
Mean dose tramadol at end of study 210 ± 113 mg/day

Tricyclics and antiepileptics discontinued ≥ 21 days; shorter acting analgesics discontinued ≥ 7 days before start

Rescue medication: "No pain medications other than the study medications were permitted"

Outcomes

PI at end of study (5‐point scale, 0‐4)
PR (6‐point scale, ‐1 to 4)

Adverse events

Notes

USA. Sponsor: Ortho‐McNeil Pharmaceutical, Raritan, NJ (research grant)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer random‐number generator

Allocation concealment (selection bias)

Low risk

Double‐blind code numbers assigned sequentially

Blinding (performance bias and detection bias)
All outcomes

Low risk

Identically appearing capsules, indistinguishable

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Imputation not mentioned, approximately 30% withdrawals, with different reasons between groups

Size

Unclear risk

50‐199 participants per treatment arm (65, 66)

Norrbrink 2009

Methods

Randomised, double‐blind, placebo‐controlled, parallel‐group

Duration: 4 weeks

Assessed at baseline and week 4 (daily pain diary)

Participants

Spinal cord injury ≥ 12 months, "at or below level of lesion neuropathic pain" ≥ 6 months, PI > 3 (Borg's Category Ratio), aged 18‐70 years
Exclusion: cognitive impairment; previous treatment with tramadol; intolerance to opioids in past

Current use of opioids or antidepressants considered on individual basis

N = 35
M 28, F 7
Mean age 51 years (SD 11)
Mean 15 years post injury
Some differences in baseline characteristics ‐ level of injury, baseline PI

Worst PI at baseline: 7‐9/10, but general PI 4‐7/10

Interventions

Tramadol 50 mg x 3 daily, n = 23
Placebo, n = 12

Dose increased every 5 days by 50 mg (1 tablet) to maximum of 400 mg/d (or 8 placebo tablets) until optimal pain relief or intolerable adverse events

Stable pain medication allowed without change to dosage (20/35 took concomitant pain medication)

Outcomes

Daily PI: complete relief = 10
PGIC
Adverse events

Notes

Sweden. Sponsor: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method not described

Allocation concealment (selection bias)

Low risk

"sealed coded envelopes" provided by third party

Blinding (performance bias and detection bias)
All outcomes

Low risk

"identical in appearance"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT analysis with LOCF

Size

High risk

< 50 participants per treatment arm (12, 23)

Sindrup 1999

Methods

Randomised, double‐blind, placebo‐controlled, cross‐over

Duration: 2 x 4 weeks with washout of ≥ 1 week between periods

Assessed at end of treatment periods

Participants

Polyneuropathy > 6 months, PI without treatment ≥ 4/10, aged 20‐80 years
Exclusion: pain from other causes; previous allergy to tramadol; intolerance to tramadol or other opioids; use of MAO inhibitors; epilepsy; severe terminal illness

N = 45 (34 provided data for both periods)

M 27, F 18

Median age 58 years (range 26‐77)

Median baseline PI: 6/10

Interventions

Tramadol SR, titrated to 100‐200 mg twice daily over at least 1 week
Placebo

(22 participants took tramadol first, 23 placebo first)

Rescue medication: up to 6 x paracetamol 500 mg/d

Existing pain medication slowly discontinued over a maximum of 1 week

Outcomes

Daily PI: NRS 0‐10 (also paraesthesia and touch‐evoked pain) used to calculate median for each week
Use of rescue medication
Adverse events
Preference at end of study

Notes

Denmark. Sponsor: Grunenthal GmbH

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"computer generated randomisation code with a block size of six"

Allocation concealment (selection bias)

Low risk

"sealed envelopes", participants numbered consecutively and treated with drugs with corresponding randomisation number

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Method not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

LOCF imputation, efficacy data only for participants providing data for both phases; reasons for withdrawals per treatment arm not fully reported

Size

High risk

< 50 participants per treatment arm (≤ 43, 40)

Sindrup 2012

Methods

Multicentre, randomised, double‐blind, active‐ and placebo‐controlled, cross‐over

Duration: 3 x 4 weeks with washout of 1‐2 weeks between periods

Assessed weekly and at end of each treatment period

Participants

Polyneuropathy (distal, symmetric) > 6 months, PI ≥ 4/10, aged 18‐74 years
Exclusion: pain from other causes; psychiatric disease; raised creatinine or liver enzymes; chronic disease affecting drug absorption; HbA1c > 12%; QT interval > 500 ms; contraindication to opioids, tramadol, paracetamol; Hx drug or alcohol abuse; use of other analgesics or MAO inhibitors or non‐pharmacological pain therapy

N = 64 (48 completed)
M 44, F 20
Mean age 58 years (38‐75)

Baseline PI: 6/10

Interventions

Tramadol SR 100 mg/d, increasing to 200‐400 mg/d
GRT9906 (experimental drug) 60 mg/d, increasing to 120‐240 mg/d
Placebo

Dose increased over 1 week, then kept constant for remaining 3 weeks
Most participants took maximum dose

Rescue medication: paracetamol up to 6 x 500 mg/d

Outcomes

Daily PI: (NRS 0‐10), then averaged over last 3 days of each period, and for each week
≥ 50% and ≥ 30% reduction in pain at end of each period

PGIC (7‐point scale; 1 = very much better)
Use of rescue medication
Adverse events

Notes

Denmark, Germany. Sponsor: Grunenthal GmbH

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"the randomization list was generated via computer"

Allocation concealment (selection bias)

Low risk

Each site given "a unique series of numbers which were assigned to each trial patient in ascending order and marked at the corresponding drug packages"

Blinding (performance bias and detection bias)
All outcomes

Low risk

The three treatments "had identical appearance and weight and were dosed similarly"

Incomplete outcome data (attrition bias)
All outcomes

High risk

Completer analysis used for PP analysis of dichotomous efficacy data.

Size

High risk

< 50 participants per treatment arm for efficacy data (maximum 56 for safety data)

F: female; HbA1c: glycosylated haemoglobin; Hx: history of; LOCF: last observation carried forward; M: male; MAO: monoamine oxidase; N: number of participants in study; n: number of participants in treatment arm; NRS: numerical rating scale; PGIC: Patient Global Impression of Change; PI: pain intensity; PP: per protocol; PR: pain relief; SD: standard deviation; SR: sustained‐release; VAS: visual analogue scale; VRS: verbal rating scale.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Ashry 2001

Commentary on Harati 1998

Attal 2001

Review article

Benedetti 1998

Investigates buprenorphine, not tramadol

Erdine 1997

Short conference abstract with inadequate method description and no usable data

Gobel 1995

Open‐label study

Harati 1999

Correspondence with no new trial data

Harati 2000

Follow up to Harati 1998. Not randomised or controlled

Herrera Silva 2001

Review article

Leppert 2001

Open‐label study

Moulin 1999

Correspondence with no new trial data

NCT00610155

7‐day treatment periods, < 10 participants per treatment arm

Saxena 2013

Open‐label cohort study

Xiao 2004

Observational study. Not a randomised controlled trial

Characteristics of studies awaiting assessment [ordered by study ID]

Ho 2009

Methods

Enriched enrolment, randomised withdrawal design. Single‐blind run‐in phases in which participants were treated with gabapentin then active placebo. Responders were randomised to a double‐blind, 3‐period cross‐over of gabapentin, tramadol, and active placebo
Participants with PI ≤ 7.5/10 at end of Period A could proceed to Period B
Participants whose average pain scores at end of Period B were ≥ 3 and increased by ≥ 30% from Period A could proceed to randomisation and a double‐blind cross‐over phase

Duration: Period A: 1 week; Period B: 2 weeks; double‐blind: 3 x 2‐week periods (1‐week titration, 1‐week stable), each followed by 1‐week washout

Participants

Idiopathic small fibre neuropathy ≥ 2 months, self‐reported gabapentin responders (on stable dose 900 ‐ 4800 mg/d), PI > 3 to ≤ 7.5 on medication, aged ≥ 18 years
Exclusion: allergies to any study drug; Hx fibromyalgia, epilepsy; cancer within 5 years; pernicious anaemia; HIV infection; multi‐organ autoimmune disease; peripheral vascular disease; renal or hepatic disease; use of insulin or antiglycaemic drugs

N = 59 entered run‐in A, 48 entered run‐in B
M 21, F 20
Mean age 60 years
N = 18 randomised
M 10, F 8
Mean age 59 years
Baseline PI: 4.9/10
Baseline PGIC 5.5 (after B)

Interventions

Period A: gabapentin at pre‐study dose + matching active placebo (diphenhydramine). Pain scores ≤ 7.5/10 entered period B
Period B: gabapentin at pre‐study dose + matching active placebo (diphenhydramine) with tapering off gabapentin. Pain scores ≥ 3/10 and increasing by ≥ 30% entered treatment period

Treatment period (2‐week test and 1‐week washout in multiple cross‐overs):
Tramadol 50 mg x 4 daily
Gabapentin pre‐study dose
Placebo (diphenhydramine 50 mg at bedtime)

Rescue medication: 325 mg tablets (probably paracetamol) ‐ limit not specified
If still inadequate, additional 400 mg gabapentin every 8 h, up to 1200 every 24 h

Outcomes

Daily PI: (NRS 0‐10), averaged over 24 h. If rescue medication or additional gabapentin taken, used score before first rescue dose of the day
PGIC (7‐point scale; 1 = very much better)
Adverse events

Notes

USA. Sponsor: Merck Research Laboratories

F: female; Hx: history of; M: male; N: number of participants in study; NRS: numerical rating scale; PGIC: Patient Global Impression of Change; PI: pain intensity.

Data and analyses

Open in table viewer
Comparison 1. Tramadol versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants with ≥ 50% pain intensity reduction Show forest plot

3

265

Risk Ratio (M‐H, Random, 95% CI)

2.16 [1.02, 4.58]

Analysis 1.1

Comparison 1 Tramadol versus placebo, Outcome 1 Participants with ≥ 50% pain intensity reduction.

Comparison 1 Tramadol versus placebo, Outcome 1 Participants with ≥ 50% pain intensity reduction.

2 Withdrawal due to adverse events Show forest plot

6

485

Risk Ratio (M‐H, Fixed, 95% CI)

4.08 [1.99, 8.37]

Analysis 1.2

Comparison 1 Tramadol versus placebo, Outcome 2 Withdrawal due to adverse events.

Comparison 1 Tramadol versus placebo, Outcome 2 Withdrawal due to adverse events.

3 All cause withdrawal Show forest plot

3

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [0.75, 1.76]

Analysis 1.3

Comparison 1 Tramadol versus placebo, Outcome 3 All cause withdrawal.

Comparison 1 Tramadol versus placebo, Outcome 3 All cause withdrawal.

4 Participants with any adverse event Show forest plot

4

266

Risk Ratio (M‐H, Fixed, 95% CI)

1.61 [1.22, 2.13]

Analysis 1.4

Comparison 1 Tramadol versus placebo, Outcome 4 Participants with any adverse event.

Comparison 1 Tramadol versus placebo, Outcome 4 Participants with any adverse event.

5 Participants with specific adverse events Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 Tramadol versus placebo, Outcome 5 Participants with specific adverse events.

Comparison 1 Tramadol versus placebo, Outcome 5 Participants with specific adverse events.

5.1 Nausea

6

508

Risk Ratio (M‐H, Fixed, 95% CI)

3.62 [2.23, 5.88]

5.2 Constipation

5

381

Risk Ratio (M‐H, Fixed, 95% CI)

4.11 [2.36, 7.16]

5.3 Tiredness/fatigue/somnolence

4

345

Risk Ratio (M‐H, Fixed, 95% CI)

3.22 [1.93, 5.36]

5.4 Dizziness

3

214

Risk Ratio (M‐H, Fixed, 95% CI)

3.72 [1.94, 7.12]

5.5 Dry mouth

3

214

Risk Ratio (M‐H, Fixed, 95% CI)

2.44 [1.35, 4.42]

Study flow diagram for the updated search
Figuras y tablas -
Figure 1

Study flow diagram for the updated search

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

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

Forest plot of comparison: 1 Tramadol versus placebo, outcome: 1.1 Participants with ≥ 50% pain intensity reduction.
Figuras y tablas -
Figure 3

Forest plot of comparison: 1 Tramadol versus placebo, outcome: 1.1 Participants with ≥ 50% pain intensity reduction.

Comparison 1 Tramadol versus placebo, Outcome 1 Participants with ≥ 50% pain intensity reduction.
Figuras y tablas -
Analysis 1.1

Comparison 1 Tramadol versus placebo, Outcome 1 Participants with ≥ 50% pain intensity reduction.

Comparison 1 Tramadol versus placebo, Outcome 2 Withdrawal due to adverse events.
Figuras y tablas -
Analysis 1.2

Comparison 1 Tramadol versus placebo, Outcome 2 Withdrawal due to adverse events.

Comparison 1 Tramadol versus placebo, Outcome 3 All cause withdrawal.
Figuras y tablas -
Analysis 1.3

Comparison 1 Tramadol versus placebo, Outcome 3 All cause withdrawal.

Comparison 1 Tramadol versus placebo, Outcome 4 Participants with any adverse event.
Figuras y tablas -
Analysis 1.4

Comparison 1 Tramadol versus placebo, Outcome 4 Participants with any adverse event.

Comparison 1 Tramadol versus placebo, Outcome 5 Participants with specific adverse events.
Figuras y tablas -
Analysis 1.5

Comparison 1 Tramadol versus placebo, Outcome 5 Participants with specific adverse events.

Summary of findings for the main comparison. Tramadol compared with placebo for neuropathic pain

Tramadol compared with placebo for neuropathic pain

Patient or population: adults with neuropathic pain (any origin)

Settings: community

Intervention: oral tramadol (typically started at a dose of about 100 mg daily and increased over 1 to 2 weeks to a maximum of 400 mg daily)

Comparison: placebo

Outcomes

(at trial end)

Probable outcome with
tramadol

Probable outcome with
placebo

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

At least 30% reduction in pain

Not analysed

Not analysed

Not analysed

157 participants

(2 studies)

60 events

Low quality1

At least 50% reduction in pain

530 per 1000

300 per 1000

RR 2.2 (1.02, 4.6)

NNT 4.4 (2.9 to 8.8)

265 participants

(3 studies)

110 events

Low quality1

PGIC much or very much improved

Not analysed

Not analysed

Not analysed

35 participants

(1 study)

4 events

Very low quality2

Withdrawal due to adverse event

160 per 100

30 per 1000

RR 4.1 (2.0 to 8.4)

NNH 8.2 (5.8 to 14)

485 participants

(6 studies)

45 events

Low quality1

Participants experiencing any adverse event

580 per 1000

340 per 1000

RR 1.6 (1.2 to 2.1)

NNH 4.2 (2.8 to 8.3)

266 participants

(4 studies)

123 events

Low quality1

Serious adverse events

4 serious adverse events reported in total

Not all studies reported specifically on serious adverse events

Very low quality2

Death

No data

No data

Not calculated

No data

Very low quality3

CI: confidence interval; NNH: number needed to treat for one additional harmful outcome; PGIC: Patient Global Impression of Change; RR: risk ratio

Descriptors for levels of evidence (EPOC 2015):
High quality: this research provides a very good indication of the likely effect. The likelihood that the effect will be substantially different is low.
Moderate quality: this research provides a good indication of the likely effect. The likelihood that the effect will be substantially different is moderate.
Low quality: this research provides some indication of the likely effect. However, the likelihood that it will be substantially different is high.
Very low quality: this research does not provide a reliable indication of the likely effect. The likelihood that the effect will be substantially different is very high.

Substantially different: a large enough difference that it might affect a decision.

1Downgraded 2 levels due to small number of studies and participants and relatively few events, and several sources of potential bias.
2Downgraded 3 levels due to small number of studies, and participants and events, and several sources of potential bias.
3No events.

Figuras y tablas -
Summary of findings for the main comparison. Tramadol compared with placebo for neuropathic pain
Comparison 1. Tramadol versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants with ≥ 50% pain intensity reduction Show forest plot

3

265

Risk Ratio (M‐H, Random, 95% CI)

2.16 [1.02, 4.58]

2 Withdrawal due to adverse events Show forest plot

6

485

Risk Ratio (M‐H, Fixed, 95% CI)

4.08 [1.99, 8.37]

3 All cause withdrawal Show forest plot

3

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [0.75, 1.76]

4 Participants with any adverse event Show forest plot

4

266

Risk Ratio (M‐H, Fixed, 95% CI)

1.61 [1.22, 2.13]

5 Participants with specific adverse events Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

5.1 Nausea

6

508

Risk Ratio (M‐H, Fixed, 95% CI)

3.62 [2.23, 5.88]

5.2 Constipation

5

381

Risk Ratio (M‐H, Fixed, 95% CI)

4.11 [2.36, 7.16]

5.3 Tiredness/fatigue/somnolence

4

345

Risk Ratio (M‐H, Fixed, 95% CI)

3.22 [1.93, 5.36]

5.4 Dizziness

3

214

Risk Ratio (M‐H, Fixed, 95% CI)

3.72 [1.94, 7.12]

5.5 Dry mouth

3

214

Risk Ratio (M‐H, Fixed, 95% CI)

2.44 [1.35, 4.42]

Figuras y tablas -
Comparison 1. Tramadol versus placebo