Scolaris Content Display Scolaris Content Display

Pharmakologische Interventionen zur Behandlung von Phantomschmerzen

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Abraham 2003 {published data only}

Abraham R, Marouani N, Weinbroum A. Dextromethorphan mitigates phantom pain in cancer amputees. Annals of Surgical Oncology 2003;10(3):268-74. CENTRAL

Bone 2002 {published data only}

Bone M, Critchley P, Buggy D. Gabapentin in postamputation phantom limb pain: a randomized, double-blind, placebo-controlled, cross-over study. Regional Anesthesia and Pain Medicine 2002;27(5):481-6. CENTRAL

Casale 2009 {published data only}

Casale R, Ceccherelli F, Labeeb A, Biella G. Phantom limb pain relief by contralateral myofascial injection with local anaesthetic in a placebo-controlled study: preliminary results. Journal of Rehabilitation Medicine 2009;41(6):418-22. CENTRAL

Eichenberger 2008 {published data only}

Eichenberger U, Neff F, Sveticic G, Bjorgo S, Petersen-Felix S, Arendt-Nielsen L, et al. Chronic phantom limb pain: the effects of calcitonin, ketamine, and their combination on pain and sensory thresholds. Anesthesia & Analgesia 2008;106(4):1265-73. CENTRAL

Huse 2001 {published data only}

Huse E, Larbig W, Flor H, Birbaumer N. The effect of opioids on phantom limb pain and cortical reorganization. Pain 2001;90(1-2):47-55. CENTRAL

Jaeger 1992 {published data only}

Jaeger H, Maier C. Calcitonin in phantom limb pain: a double-blind study. Pain 1992;48(1):21-7. CENTRAL

Maier 2003 {published data only}

Maier C, Dertwinkel R, Mansourian N, Hosbach I, Schwenkreis P, Senne I, et al. Efficacy of the NMDA-receptor antagonist memantine in patients with chronic phantom limb pain - results of a randomized double-blinded, placebo-controlled trial. Pain 2003;103(3):277-83. CENTRAL

Nikolajsen 1996 {published data only}

Nikolajsen L, Hansen C, Nielsen J, Keller J, Arendt-Nielsen L, Jensen T. The effect of ketamine on phantom pain: a central neuropathic disorder maintained by peripheral input. Pain 1996;67(1):69-77. CENTRAL

Robinson 2004 {published data only}

Robinson L, Czerniecki J, Ehde D, Edwards T, Judish D, Goldberg M, et al. Trial of amitriptyline for relief of pain in amputees: results of a randomized controlled study. Archives of Physical Medicine and Rehabilitation 2004;85(1):1-6. CENTRAL

Schwenkreis 2003 {published data only}

Schwenkreis P, Maier C, Pleger B, Mansourian N, Dertwinkel R, Malin J-P, et al. NMDA-mediated mechanisms in cortical excitability changes after limb amputation. Acta Neurologica Scandinavica 2003;108(3):179-84. CENTRAL

Smith 2005 {published data only}

Smith D, Ehde D, Hanley M, Campbell K, Jensen M, Hoffman A, et al. Efficacy of gabapentin in treating chronic phantom limb and residual limb pain. Journal of Rehabilitation Research & Development 2005;42(5):645-54. CENTRAL

Wiech 2004 {published data only}

Wiech K, Kiefer RT, Topfner S, Preissl H, Braun C, Unertl K, et al. A placebo-controlled randomized crossover trial of the N-methyl-D-aspartic acid receptor antagonist, memantine, in patients with chronic phantom limb pain. Anesthesia & Analgesia 2004;98(2):408-13. CENTRAL

Wu 2002 {published data only}

Wu C, Tella P, Staats P, Vaslav R, Kazim D, Wesselmann U, et al. Analgesic effects of intravenous lidocaine and morphine on postamputation pain. Anesthesiology 2002;96(2):841-8. CENTRAL

Wu 2012 {published data only}

Wu H, Sultana R, Taylor KB, Szabo A. A prospective randomized double-blinded pilot study to examine the effect of botulinum toxin type A injection versus Lidocaine/Depomedrol injection on residual and phantom limb pain: initial report. Clinical Journal of Pain 2012;28(2):108-12. CENTRAL

References to studies excluded from this review

Abraham 2002 {published data only}

Abraham R, Marouani N, Kollender Y, Meller I, Weinbroum A. Dextromethorphan for phantom pain attenuation in cancer amputees: a double-blind crossover trial involving three patients. Clinical Journal of Pain 2002;18(5):282-5. CENTRAL

Atesalp 2000 {published data only}

Ateşalp AS, Özkan Y, Kömürcü M, Erler K, Işımer A, Güret E. The effects of capsaicin in phantom limb pain [Fantom ekstremite ağrısında kapsaisinin etkileri]. Agri 2000;12(2):30-3. CENTRAL

Borghi 2010 {published data only}

Borghi B, D'Addabbo M, White PF, Gallerani P, Toccaceli L, Raffaeli W, et al. The use of prolonged peripheral neural blockade after lower extremity amputation: the effect on symptoms associated with phantom limb syndrome. Anesthesia & Analgesia 2010;111(5):1308-15. CENTRAL

Cohen 2011 {published data only}

Cohen SP, Gambel JM, Raja SN, Galvagno S. The contribution of sympathetic mechanisms to postamputation phantom and residual limb pain: a pilot study. Journal of Pain 2011;12(8):859-67. CENTRAL

Elizaga 1994 {published data only}

Elizaga AM, Smith DG, Sharar SR, Edwards WT, Hansen ST Jr. Continuous regional analgesia by intraneural block: effect on postoperative opioid requirements and phantom limb pain following amputation. Journal of Rehabilitation and Development 1994;31(3):179-87. CENTRAL

Elrazek 2005 {published data only}

Elrazek EA. The analgesic effects of epidural diamorphine and levobupivacaine on established lower limb post-amputation stump pain - a comparative study. Middle East Journal of Anesthesiology  2005;18(1):149-60. CENTRAL

Grant 2008 {published data only}

Grant AJ, Wood C. The effect of intra-neural local anaesthetic infusion on pain following major lower limb amputation. Scottish Medical Journal 2008;53(1):4-6. CENTRAL

Ilfeld 2013 {published data only}

Ilfeld BM, Moeller‐Bertram T, Hanling SR, Tokarz K, Mariano ER, Loland VJ, et al. Treating intractable phantom limb pain with ambulatory continuous peripheral nerve blocks: a pilot study. Pain Medicine 2013;14(6):935-42. CENTRAL

Jacobson 1989 {published data only}

Jacobson L, Chabala C, Brody M. Relief of persistent postamputation stump and phantom limb pain with intrathecal fentanyl. Pain  1989;37(3):317-22. CENTRAL

Jacobson 1990 {published data only}

Jacobson L, Chabal C, Brody MC, Mariano AJ, Chaney EF. A comparison of the effects of intrathecal fentanyl and lidocaine on established postamputation stump pain. Pain 190;40(2):137-41. CENTRAL

Jaeger 1988 {published data only}

Jaeger H, Maier C, Wawersik J. Postoperative treatment of phantom pain and causalgias with calcitonin. Anaesthesist  1988;37(2):71-7. CENTRAL

Jin 2009 {published data only}

Jin L, Kollewe K,  Krampfl K, Dengler R, Mohammadi B. Treatment of phantom limb pain with botulinum toxin type A. Pain Medicine 2009;10(2):300-3. CENTRAL

Karanikolas 2011 {published data only}

Karanikolas M, Aretha D, Tsolakis I, Monantera G, Kiekkas P, Papadoulas S, et al. Optimized perioperative analgesia reduces chronic phantom limb pain intensity, prevalence, and frequency: a prospective, randomized, clinical trial. Anesthesiology 2011;114(5):1144-54. CENTRAL

Kessel 1987 {published data only}

Kessel C, Wörz R. Immediate response of phantom limb pain to calcitonin. Pain 1987;30(1):79-87. CENTRAL

Kukushkin 1996 {published data only}

Kukushkin ML, Ivanova AF, Ovechkin AM, Gnezdilov AV, Reshetniak VK. Differential combined drug therapy of phantom pain syndrome after amputation of extremity. Anesteziologiia i Reanimatologiia 1996;4:39-42. CENTRAL

Licina 2013 {published data only}

Licina L, Hamsher C, Lautenschager K, Dhanjal S, Williams N, Spevak C. Buprenorphine/naloxone therapy for opioid refractory neuropathic pain following traumatic amputation: a case series. Military Medicine 2013;178(7):e858-61. CENTRAL

Lirk 2012 {published data only}

Lirk P, Stadlbauer KH, Hollmann MW. ESA Clinical Trials Network 2012: PLATA – Prevention of Phantom Limb Pain After Transtibial Amputation: Randomised, double-blind, controlled, multicentre trial comparing optimised intravenous pain control versus optimised intravenous pain control plus regional anaesthesia. European Journal of Anaesthesiology 2013;30(5):202-4. CENTRAL

Neil 2012 {published data only}

Neil MJ. Brachial plexus block in phantom limb pain: interesting, but limited clinical benefit in chronic setting. Pain Medicine 2012;13(6):849. CENTRAL

Nikolajsen 1997 {published data only}

Nikolajsen L, Hansen PO, Jensen TS. Oral ketamine therapy in the treatment of postamputation stump pain. Acta Anaesthesiologica Scandinavica 1997;41(3):427-9. CENTRAL

Nikolajsen 2006 {published data only}

Nikolajsen L, Finnerup NB, Kramp S, Vimtrup AS, Keller J, Jensen TS. A randomized study of the effects of gabapentin on postamputation pain. Anesthesiology 2006;105(5):1008-15. CENTRAL

Panerai 1990 {published data only}

Panerai AE, Monza G, Movilia P, Bianchi M, Francucci BM, Tiengo M. A randomized, within-patient, cross-over, placebo-controlled trial on the efficacy and tolerability of the tricyclic antidepressants chlorimipramine and nortriptyline in central pain. Acta Neurologica Scandinavica 1990;82(1):34-8. CENTRAL

Pinzur 1996 {published data only}

Pinzur MS, Garla PG, Pluth T, Vrbos L. Continuous postoperative infusion of a regional anesthetic after an amputation of the lower extremity. A randomized clinical trial. Journal of Bone and Joint Surgery. American volume 1996;78(10):1501-5. CENTRAL

Rogers 1989 {published data only}

Rogers AG. Use of amitriptyline (Elavil) for phantom limb pain in younger children. Journal of Pain & Symptom Management  1989;4(2):96. CENTRAL

Sato 2008 {published data only}

Sato K, Higuchi H, Hishikawa Y. Management of phantom limb pain and sensation with milnacipran. Journal of Neuropsychiatry & Clinical Neurosciences 2008;20(3):368. CENTRAL

Scadding 1982 {published data only}

Scadding JW, Wall PD, Parry CB, Brooks DM. Clinical trial of propranolol in post-traumatic neuralgia. Pain 1982;14(3):283-92. CENTRAL

Van Seventer 2010 {published data only}

Van Seventer R, Bach FW, Toth CC, Serpell M, Temple J, Murphy TK, et al. Pregabalin in the treatment of post-traumatic peripheral neuropathic pain: a randomized double-blind trial. European Journal of Neurology 2010;17(8):1082-9. CENTRAL

Vorobeichik 1997 {published data only}

Vorobeĭchik IaM, Kukushkin ML, Reshetniak VK, Ovechkin AM, Gnezdilov AV. The treatment of the phantom pain syndrome with tizanidine. Zh Nevrol Psikhiatr Im S S Korsakova 1997;97(3):36-9. CENTRAL

Wilder‐Smith 2005 {published data only}

Wilder-Smith CH, Hill LT, Laurent S. Postamputation pain and sensory changes in treatment-naive patients: characteristics and responses to treatment with tramadol, amitriptyline, and placebo. Anesthesiology 2005;103(3):619-28. CENTRAL

Wu 2008 {published data only}

Wu CL, Agarwal S, Tella PK, Klick B, Clark MR, Haythornthwaite JA, et al. Morphine versus mexiletine for treatment of postamputation pain: a randomized, placebo-controlled, crossover trial. Anesthesiology 2008;109(2):289-96. CENTRAL

References to studies awaiting assessment

Buch 2019 {published data only}

Buch NS, Ahlburg P, Haroutounian S, Anderson NT, Finnerup NB, Nikolajsen L. The role of afferent input in postamputation pain: a randomized, double-blind, placebo-controlled crossover study. Pain 2019;160(7):1622-33. CENTRAL [DOI: 10.1097/j.pain.0000000000001536]

Aoki 2001a

Aoki KR. Pharmacology and immunology of botulinum toxin serotypes. Journal of Neurology 2001;248(1):I3-10.

Aoki 2001b

Aoki KR, Guyer B. Botulinum toxin type A and other botulinum toxin serotypes: a comparative review of biochemical and pharmacological actions. European Journal of Neurology 2001;8(s5):21-9.

Azria 2002

Azria M. Possible mechanisms of the analgesic action of calcitonin. Bone 2002;30(5):80S-3S.

Bileviciute‐Ljungara 2001

Bileviciute-Ljungara I, Biellab G, Bellomib P, Sotgiub M. Contralateral treatment with lidocaine reduces spinal neuronal activity in mononeuropathic rat. Neuroscience Letters 2001;311:157-60.

Birbaumer 1997

Birbaumer N, Lutzenberger W, Montoya P, Larbig W, Unertl K, Topfner S, et al. Effects of regional anesthesia on phantom limb pain are mirrored in changes in cortical reorganization. Journal of Neuroscience 1997;17(14):5503-8.

Bolognini 2013

Bolognini N, Olgiati E, Maravita A, Ferraro F, Fregni F. Motor and parietal cortex stimulation for phantom limb pain and sensations. Pain 2013;154(8):1274-80.

Brin 1997

Brin MF. Botulinum toxin: chemistry, pharmacology, toxicity, and immunology. Muscle Nerve Supplement 1997;6:S146-68.

Burgoyne 2012

Burgoyne L, Billups C, Jirón J, Kaddoum R, Wright B, Bikhazi G, et al. Phantom limb pain in young cancer-related amputees: recent experience at St. Jude Children’s Research Hospital. Clinical Journal of Pain 2012;28(3):222-5.

Charrow 2008

Charrow A, DiFazio M, Foster L, Pasquina PF, Tsao JW. Intradermal botulinum toxin type A injection effectively reduces residual limb hyperhidrosis in amputees: a case series. Archives of Physical Medicine and Rehabilitation 2008;89(7):1407-9.

Clark 2013

Clark RL, Bowling FL, Jepson F, Rajbhandari S. Phantom limb pain after amputation in diabetic patients does not differ from that after amputation in nondiabetic patients. Pain 2013;154(5):729-32.

Darnall 2005

Darnall BD, Ephraim P, Wegener ST, Dillingham T, Pezzin L, Rossbach P, et al. Depressive symptoms and mental health service utilization among persons with limb loss: results of a national survey. Archives of Physical Medicine and Rehabilitation 2005;86(4):650-8.

Desmond 2010

Desmond DM, MacLachlan M. Prevalence and characteristics of phantom limb pain and residual limb pain in the long term after upper limb amputation. International Journal of Rehabilitation Research 2010;33(3):279-82.

Ehde 2000

Ehde DM, Czerniecki JM, Smith DG, Campbell KM, Edwards WT, Jensen MP, et al. Chronic phantom sensations, phantom pain, residual limb pain, and other regional pain after lower limb amputation. Archives of Physical Medicine and Rehabilitation 2000;81(8):1039-44.

Eisenberg 1998

Eisenberg E, Kleiser A, Dortort A, Haim T, Yarnitsky D. The NMDA (N-methyl-D-aspartate) receptor antagonist memantine in the treatment of postherpetic neuralgia: a double-blind, placebo-controlled study. European Journal of Pain 1998;2(4):321-7.

Elbert 1994

Elbert T, Flor H, Birbaumer N, Knecht S, Hampson S, Larbig W, et al. Extensive reorganization of the somatosensory cortex in adult humans after nervous system injury. NeuroReport 1994;5(18):2593-7.

Elbert 2004

Elbert T, Rockstroh B. Reorganization of human cerebral cortex: the range of changes following use and injury. Neuroscientist 2004;10(2):129-41.

Ephraim 2005

Ephraim PL, Wegener ST, MacKenzie EJ, Dillingham TR, Pezzin LE. Phantom pain, residual limb pain, and back pain in amputees: results of a national survey. Archives of Physical Medicine and Rehabilitation 2005;86(10):1910-9.

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.

Flor 1995

Flor H, Elbert T, Knecht S, Wienbruch C, Pantev C, Birbaumer N, et al. Phantom limb pain as a perceptual correlate of cortical reorganization following arm amputation. Nature 1995;375(6531):482-4.

Foell 2011

Foell J, Bekrater-Bodmann R, Flor H, Cole J. Phantom limb pain after lower limb trauma origins and treatments. International Journal of Lower Extremity Wounds 2011;10(4):224-35.

Furlan 2006

Furlan A, Sandoval J, Mailis Gagnon A, Tunks E. Opioids for chronic noncancer pain: a meta-analysis of effectiveness and side effects. Canadian Medical Association Journal 2006;174(11):1589-94.

Furlan 2011

Furlan AD, Chaparro LE, Irvin E, Mailis-Gagnon A. A comparison between enriched and nonenriched enrollment randomized withdrawal trials of opioids for chronic noncancer pain. Pain Research & Management: The Journal of the Canadian Pain Society 2011;16(5):337-51.

Giummarra 2011

Giummarra MJ, Moseley GL. Phantom limb pain and bodily awareness: current concepts and future directions. Current Opinion in Anesthesiology 2011;24(5):524-31.

Halbert 2002

Halbert J, Crotty M, Cameron ID. Evidence for the optimal management of acute and chronic phantom pain: a systematic review. Clinical Journal of Pain 2002;18(2):84-92.

Hall 2013

Hall GC, Morant, SV, Carroll D, Gabriel ZL, McQuay HJ. An observational descriptive study of the epidemiology and treatment of neuropathic pain in a UK general population. BMC Family Practice 2013;14(1):28.

Hanley 2004

Hanley MA, Jensen MP, Ehde DM, Hoffman AJ, Patterson DR, Robinson LR. Psychosocial predictors of long-term adjustment to lower-limb amputation and phantom limb pain. Disability & Rehabilitation 2004;26(14-15):882-93.

Hanley 2009

Hanley MA, Ehde DM, Jensen M, Czerniecki J, Smith DG, Robinson LR. Chronic pain associated with upper-limb loss. American Journal of Physical Medicine & Rehabilitation 2009;88(9):742-79.

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 handbook.cochrane.org.

Jensen 1983

Jensen TS, Krebs B, Nielsen J, Rasmussen P. Phantom limb, phantom pain and stump pain in amputees during the first 6 months following limb amputation. Pain 1983;17(3):243-56.

Jensen 1985

Jensen TS, Krebs B, Nielsen J, Rasmussen P. Immediate and long-term phantom limb pain in amputees: incidence, clinical characteristics and relationship to pre-amputation limb pain. Pain 1985;21(3):267-8.

Jensen 2000

Jensen TS, Nikolajsen L. Pre-emptive analgesia in postamputation pain: an update. Progress in Brain Research 2000;129:493-503.

Kalso 2004

Kalso E, Edwards J, Moore A, McQuay H. Opioids in chronic non-cancer pain: systematic review of efficacy and safety. Pain 2004;112(3):372-80.

Katz 1990

Katz J, Melzack R. Pain “memories” in phantom limbs: review and clinical observations. Pain 1990;43(3):319-36.

Kern 2003

Kern U, Martin C, Scheicher S, Müller H. Treatment of phantom pain with botulinum-toxin A. A pilot study [Abstract]. Schmerz 2003;17(2):117-24.

Kern 2004a

Kern U, Martin C, Scheicher S, Muller H. Does botulinum toxin A make prosthesis use easier for amputees? Journal of Rehabilitation Medicine 2004;36(5):238-9.

Kern 2004b

Kern U, Martin C, Scheicher S, Müller H. Long-term treatment of phantom and stump pain with Botulinum toxin type A over 12 months. A first clinical observation [Abstract]. Der Nervenarzt 2004;75(4):336-40.

Kern 2011

Kern U, Kohl M, Seifert U, Schlereth T. Botulinum toxin type B in the treatment of residual limb hyperhidrosis for lower limb amputees: a pilot study. American Journal of Physical Medicine & Rehabilitation 2011;90(4):321-9.

Khanahmadi 2012

Khanahmadi S, Skrabek R, Arneja A. Effect of nabilone on phantom limb pain [Conference Abstract]. Journal of Rehabilitation Medicine 2012;44(11):1004.

Lei 2004

Lei LG, Sun S, Gao YJ, Zhao ZQ, Zhang YQ. NMDA receptors in the anterior cingulated cortex mediate pain-related aversion. Experimental Neurology 2004;189(2):413-21.

Manchikanti 2004

Manchikanti L, Singh V. Managing phantom pain. Pain Physician 2004;7(3):365-75.

McCormick 2014

McCormick Z, Chang‐Chien G, Marshall B, Huang M, Harden RN. Phantom limb pain: a systematic neuroanatomical‐based review of pharmacologic treatment. Pain Medicine 2014;15(2):292-305.

McNicol 2013

McNicol ED, Midbari A, Eisenberg E. Opioids for neuropathic pain. Cochrane Database of Systematic Reviews 2013, Issue 8. Art. No: CD006146. [DOI: 10.1002/14651858.CD006146.pub2]

Millstein 1985

Millstein S, Bain D, Hunter GA. A review of employment patterns of industrial amputees - factors influencing rehabilitation. Prosthetics and Orthotics International 1985;9(2):69-78.

Montoya 1998

Montoya P, Ritter K, Huse E, Larbig W, Braun C, Töpfner S, et al. The cortical somatotopic map and phantom phenomena in subjects with congenital limb atrophy and traumatic amputees with phantom limb pain. European Journal of Neuroscience 1998;10(3):1095-102.

Moore 2005

Moore RA, McQuay HJ. Prevalence of opioid adverse events in chronic non-malignant pain: systematic review of randomised trials of oral opioids. Arthritis Research & Therapy 2005;7(5):R1046-51.

Moore 2014

Moore RA, Wiffen PJ, Derry S, Toelle T, Rice ASC. Gabapentin for chronic neuropathic pain and fibromyalgia in adults. Cochrane Database of Systematic Reviews 2014, Issue 4. Art. No: CD007938. [DOI: 10.1002/14651858.CD007938.pub3]

Moore 2015

Moore RA, Derry S, Aldington D, Cole P, Wiffen PJ. Amitriptyline for neuropathic pain in adults. Cochrane Database of Systematic Reviews 2015, Issue 7. Art. No: CD008242. [DOI: 10.1002/14651858.CD008242.pub3]

Moseley 2012

Moseley GL, Flor H. Targeting cortical representations in the treatment of chronic pain a review. Neurorehabilitation and Neural Repair 2012;26(6):646-52.

Moulin 2014

Moulin DE, Boulanger A, Clark AJ, Clarke H, Dao T, Finley GA, et al. Pharmacological management of chronic neuropathic pain: revised consensus statement from the Canadian Pain Society. Pain Research and Management 2014;19(6):328-35.

Nikolajsen 2000

Nikolajsen L, Gottrup H, Kristensen A, Jensen T. Memantine (a n-methyl-d-aspartate receptor antagonist) in the treatment of neuropathic pain after amputation or surgery: a randomized, double-blinded, cross-over study. Anesthesia & Analgesia 2000;91(4):960-6.

Nikolajsen 2001

Nikolajsen L, Jensen TS. Phantom limb pain. British Journal of Anesthesia 2001;87(1):107-16.

Penn‐Barwell 2011

Penn-Barwell JG. Outcomes in lower limb amputation following trauma: a systematic review and meta-analysis. Injury 2011;42(12):1474-9.

Pezzin 2000

Pezzin LE, Dillingham TR, MacKenzie EJ. Rehabilitation and the long-term outcomes of persons with trauma-related amputations. Archives of Physical Medicine and Rehabilitation 2000;81(3):292-300.

Preißler 2013

Preißler S, Feiler J, Dietrich C, Hofmann GO, Miltner WH, Weiss T. Gray matter changes following limb amputation with high and low intensities of phantom limb pain. Cerebral Cortex 2013;23(5):1038-48.

Ramachandran 1992

Ramachandran VS, Rogers-Ramachandran D, Stewart M. Perceptual correlates of massive cortical reorganization. Science 1992;258(5085):1159-60.

Reiber 2010

Reiber GE, McFarland LV, Hubbard S, Maynard C, Blough DK, Gambel JM, et al. Servicemembers and veterans with major traumatic limb loss from Vietnam war and OIF/OEF conflicts: Survey methods, participants, and summary findings. Journal of Rehabilitation Research and Development 2010;47(4):275-98.

Richardson 2006

Richardson C, Glenn S, Nurmikko T, Horgan M. Incidence of phantom phenomena including phantom limb pain 6 months after major lower limb amputation in patients with peripheral vascular disease. Clinical Journal of Pain 2006;22(4):353-8.

Robbins 2009

Robbins CB, Vreeman DJ, Sothmann MS, Wilson SL, Oldridge NB. A review of the long-term health outcomes associated with war-related amputation. Military Medicine 2009;174(6):588-92.

Schley 2007

Schley M, Topfner S, Wiech K, Schaller H, Konrad C, Schmelz M, et al. Continuous brachial plexus blockade in combination with the NMDA receptor antagonist memantine prevents phantom pain in acute traumatic upper limb amputees. European Journal of Pain 2007;11(3):299-308.

Sheehan 2014

Sheehan TP, Gondo GC. Impact of limb loss in the United States. Physical Medicine & Rehabilitation Clinics of North America 2014;25:9-28.

Sherman 1980

Sherman RA. Published treatments of phantom limb pain. American Journal of Physical Medicine 1980;59(5):232-44.

Sherman 1984

Sherman RA, Sherman C, Parker L. Chrononic phantom and stump pain among American veterans: results of a survey. Pain 1984;18:83-95.

Silberstein 2001

Silberstein SD. Review of botulinum toxin type A and its clinical applications in migraine headache. Expert Opinion on Pharmacotherapy 2001;2:1649-54.

Sin 2013

Sin EI, Thong SY, Poon KH. Incidence of phantom limb phenomena after lower limb amputations in a Singapore tertiary hospital. Singapore Medical Journal 2013;54(2):75-81.

Sinha 2011a

Sinha R, Van Den Heuvel WJ. A systematic literature review of quality of life in lower limb amputees. Disability and Rehabilitation 2011;33(11):883-99.

Sinha 2011b

Sinha R, van den Heuvel WJ, Arokiasamy P. Factors affecting quality of life in lower limb amputees. Prosthetics and Orthotics International 2011;35(1):90-6.

Stansbury 2008

Stansbury LG, Lalliss SJ, Branstetter JG, Bagg MR, Holcomb JB. Amputations in US military personnel in the current conflicts in Afghanistan and Iraq. Journal of Orthopaedic Trauma 2008;22(1):43-6.

Tan 2010

Tan T, Barry P, Reken S, Baker M. Guidelines: Pharmacological management of neuropathic pain in non-specialist settings: summary of NICE guidance. BMJ 2010;340(7748):707-70.

Whyte 2002

Whyte AS, Carroll LJ. A preliminary examination of the relationship between employment, pain and disability in an amputee population. Disability and Rehabilitation 2002;24(9):462-70.

Wolff 2011

Wolff A, Vanduynhoven E, Van Kleef M, Huygen F, Pope J, Mekhail N. Phantom pain. Pain Practice 2011;11(4):403-13.

Woolf 2011

Woolf CJ. Central sensitization: implications for the diagnosis and treatment of pain. Pain 2011;152(3):S2-15.

Ziegler‐Graham 2008

Ziegler-Graham K, MacKenzie EJ, Ephraim PL, Travison TG, Brookmeyer R. Estimating the prevalence of limb loss in the United States: 2005 to 2050. Archives of Physical Medicine and Rehabilitation 2008;89:42-9.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Abraham 2003

Study characteristics

Methods

Controlled clinical trial; DB followed by open phase, 3‐period, cross‐over; no wash‐out period; non‐involved doctor prepared drugs and order of administration 

Ff‐up after 10 days of each treatment period (DB phase)

Participants

Severe phantom pain for at least 1 month despite extensive pain therapy; majority of upper and lower extremity amputations of cancer aetiology, rest due to vascular and trauma; 10 participants, 5 males; mean age in yrs (SD): 50 (14); duration of phantom pain, months: 4.8

Interventions

  1. dextromethorphan 60 mg for 10 days, oral

  2. dextromethorphan 90 mg for 10 days, oral

  3. placebo

Outcomes

Number of participants with ≥ 50% pain relief on subjective pain intensity score 0 to 100;

Feeling of well‐being from 0 to 100;

Sedation score from 0 to 100;

Adverse events;

Dropouts/withdrawals

Notes

Limitations related to dosing and small sample size; n = 10

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

Physician not involved in study prepared batches of medications and order of administration

Blinding (performance bias and detection bias)
All outcomes

Low risk

Identical capsules; outcomes assessed at the medical centre acute pain service

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Outcomes assessed at the medical centre acute pain service but not clear if blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Complete outcome data for all

Selective reporting (reporting bias)

Unclear risk

Results in the DB phase reported in graphical form noting level of significance but without numerical results; the results for the specified outcomes in the methods section of the published study were reported, although not necessarily as our preferred outcomes

Other bias

High risk

no wash‐out period; carry‐over effect not addressed

Size of study

High risk

n = 10

Bone 2002

Study characteristics

Methods

Randomised, DB, cross‐over; 6 weeks each treatment arm; 1 week wash‐out period; computer‐generated randomisation

Ff‐up at 6 wks

Participants

Phantom pain of at least 6 months with pain intensity of at least 40 mm on 100‐millimetre VAS scale; majority with lower limb amputations; time since amputation is 18 months; 19 participants, 15 males; mean age, yrs (SD): 56.25 (17.5); baseline mean pain intensity (SD) (converted and presented in cm VAS by Bone 2002 study authors): treatment group 6.1 (1.8); placebo 6.7 (1.9)

Interventions

  1. gabapentin titrated in increments of 300 mg up to 2400 mg or maximal tolerable dose for 6 weeks; oral

  2. placebo

Outcomes

Change in pain intensity 100‐millimetre VAS at end of treatment week 6 vs baseline (converted and presented in cm VAS by Bone 2002 study authors);

Mean pain intensity difference at end of treatment week 6;

Categorical phantom pain intensity (0 = none, 1 = mild pain, 2 = moderate pain, 3 = severe pain) end of treatment to baseline;

Change in mood on HADS;

Change in function on BI;

Change in sleep on SIS;

Number of rescue tablets;

Adverse events;

Dropouts/withdrawals

Notes

For lifestyle indices, sample size may be too small to rule out type 2 error; used between‐group analysis for comparisons; n = 19

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation

Allocation concealment (selection bias)

Low risk

Organized remotely (hospital pharmacist)

Blinding (performance bias and detection bias)
All outcomes

Low risk

"identical coded medication bottles containing identical tablets of gabapentin and placebo"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described who assessed and if blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

"Data from these 19 patients were included in the results presented, using intention‐to‐treat analysis"

Selective reporting (reporting bias)

Low risk

All specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes

Other bias

Low risk

(+) wash‐out period; baseline VAS pain score before placebo/gabapentin not significantly different

Size of study

High risk

n = 19

Casale 2009

Study characteristics

Methods

Randomised, DB, cross‐over with 72‐hour wash‐off period; computer‐generated randomisation

Ff‐up at 60 min after injection

Participants

PLP of at least 6 months, lower extremity amputation of traumatic and vascular aetiology; 8 participants, 6 males; mean age, yrs (SD): 70.1 (7.7); baseline pain intensity on VAS from 0 no pain to 10 worst pain ever experienced, mean (SD): 7.9 (0.8) treatment group; 7.6 (0.7) control group

Interventions

  1. contralateral myofascial injection with local anaesthetic bupivacaine at 2.5 mg/mL, 1 mL, given once

  2. placebo (saline)

Outcomes

Pain intensity on VAS from 0 no pain to 10 worst pain ever experienced;

Mean difference pain intensity;

Adverse events;

Dropouts/withdrawals;

Other outcomes: phantom sensation, mirror displacement in healthy limbs

Notes

Small number of participants; n = 8; preliminary results

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation (confirmed through email correspondence with author)

Allocation concealment (selection bias)

Low risk

"saline or local anaesthetic solutions prepared in a separate room by a nurse"

Blinding (performance bias and detection bias)
All outcomes

Low risk

"Syringes of same size; an independent physician blinded to contents of syringe performed injections"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The same physician who performed the basal clinical examination blinded to the treatment, visited the patients collecting number of painful muscle areas present within 1 hr of injection. The intensity of the phantom pain was evaluated before and after treatment by means of the VAS from 0 (no pain) to 10 (worst pain)"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants accounted for

Selective reporting (reporting bias)

Low risk

All specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes

Other bias

Low risk

(+) wash‐out period; baseline pain intensity during anaesthetic and saline not significantly different

Size of study

High risk

n = 8

Eichenberger 2008

Study characteristics

Methods

Randomised, DB, cross‐over, 1 hr each treatment arm; time between infusions 48 hours; Randomisation by drawing lots by person not involved in study

Ff‐up at 30, 60 min, 48 hrs after infusion

Participants

Chronic phantom pain > 6 months' duration; upper and lower extremity amputation of vascular, traumatic, cancer, chronic pain in aetiologies, mean pain intensity ≥ 3 on 10‐centimetre VAS scale; 20 participants; 15 males; median age, yrs (range): 57 (19.3 to 72.7); mean baseline pain intensity on 10‐centimetre VAS: 4.32; duration of phantom pain, yrs: 12.41

Interventions

  1. ketamine at 0.4 mg/kg, once, 1‐hour intravenous infusion

  2. calcitonin  at 200 IU once, 1‐hour intravenous infusion

  3. combination ketamine/calcitonin at 200 IU calcitonin and 0.4 mg/kg ketamine once, 1‐hour intravenous infusion

  4. placebo (saline)

Outcomes

Number of participants with ≥ 50% pain reduction on 10‐centimetre VAS;

Change in pain intensity on 10‐centimetre VAS;

Adverse events:

Dropouts/withdrawals;

Other outcomes: basal sensory assessments

Notes

Relatively small sample size; n = 20; wide range in duration of phantom pain; ketamine alone was given to only 10 participants

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Drawing of lots

Allocation concealment (selection bias)

Low risk

Person not involved in study randomised and prepared solutions

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

"neither investigator performing experiment nor the patients were aware of the solutions infused"

"In some cases, drug‐related side effects occurred which rendered blinding of physician performing the tests and patients questionable"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"In some cases, drug‐related side effects occurred which rendered blinding of physician performing the tests and patients questionable"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Missing outcome data in 1 group, but not related to outcome

Selective reporting (reporting bias)

Low risk

All specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes

Other bias

Unclear risk

Carry‐over effect

Size of study

High risk

n = 20

Huse 2001

Study characteristics

Methods

Randomised, cross‐over, 4‐week double‐blinded phase with 1 to 2 weeks' wash‐out period; a long‐term open phase for responders to intervention; physician with no contact with participants randomised and kept code

Ff‐up: hourly for pain and adverse event; weekly during 4 weeks of DB phase; long term 6, 12 mos (open)

Participants

Phantom pain at least 3 in 10‐centimetre VAS; with upper and lower extremity amputations; 12 participants, 10 males; mean age, yrs (SD): 50.58 (14.01); mean baseline pain intensity on 10‐centimetre VAS (SD): 4.65 (1.06); time since amputation, years (SD): 16.49 (14.01)

Interventions

  1. morphine sulfate titrated from 70 mg/day up to 300 mg/day or max tolerable dose for 4 weeks, oral

  2. placebo

Outcomes

Change in pain intensity on 0‐to‐10‐centimetre VAS;

Number of participants with pain reduction of > 50% (10‐centimetre VAS);

Change in mood/depression on Self‐Rating Depression Scale;

Long‐term outcomes (6 months, 12 months): only with morphine sulfate; n = 9

Other outcomes: pain‐related self‐assessment scale; active coping and catastrophising using West Haven‐Yale Multidimensional Pain Inventory; Brief Stress Scale; psychophysical thresholds, 2‐point discrimination; attentional performance with d2 Test of Attention, magnetoencephalography

Notes

Small sample size (n = 12); cortical reorganisation results based on 3 participants (open phase)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

Physician with no contact with participants randomised and kept code.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as a double‐blind study; efforts were made to blind participants (e.g. treatment and placebo preparations were "put into exactly identical pills by the pharmacy"; identical treatment phases). The authors of the study acknowledged in their discussion that the participants were able to guess the morphine medication (but not the placebo) due to side effects, but there was no mention of feedback as to the correctness of their guess. Also, pain reduction scores with the morphine treatment were not significantly correlated with participant‐assessed treatment expectancy outcomes.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as double‐blind study; while there was mention of psychologist (not involved in giving treatment to the participant) who administered psychological assessment scales, it was unclear who assessed pain intensity and side effects

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants accounted for in analysis of outcomes

Selective reporting (reporting bias)

Low risk

All specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes.

Other bias

Low risk

(+) wash‐out period

Size of study

High risk

n = 12

Jaeger 1992

Study characteristics

Methods

Controlled clinical trial, cross‐over; 2‐hour wash‐out period; double‐blind phase, then an open phase with the intervention (s‐calcitonin) for longer‐term assessment; drawing of lots by person not involved in study

Ff‐up: short‐term: 24 hrs before and after treatment (double‐blind); long‐term: 6 mos, 1 to 2 yrs (open phase)

Participants

Phantom pain 0 to 7 days following amputation; all except one are lower limb amputations of vascular, traumatic, malignancy, and infectious aetiology, at least 3 on 0‐to‐10 numerical analogue scale; 21 participants, 12 males; median age yrs (range): 59 (20 to 78)

Interventions

  1. s‐calcitonin at 200 IU, once, 20‐minute intravenous infusion 

  2. saline

Outcomes

Change in pain intensity on 0‐to‐10 numerical analogue scale;

Number of participants with pain reduction of > 50%;

Long term (at 1 yr): number of participants with reduction of > 75%;

Adverse events;

Dropouts/withdrawals;

Other outcomes: number of phantom pain attacks, number of participants requiring second infusion for phantom pain recurrence

Notes

n = 21

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"patients with PLP exceeding 3 on NAS were randomly divided into 2 groups"

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding (performance bias and detection bias)
All outcomes

Low risk

"double‐blind"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Some missing data, as some participants did not have the second infusion, placebo, as their NAS did not exceed 3

Selective reporting (reporting bias)

Unclear risk

Numerical results for pain intensity on NAS not reported, although out in graphical form and noted significance; all specified outcomes in methods were reported, although not necessarily our preferred outcomes

Other bias

Unclear risk

Unclear if baseline pain characteristics were not significantly different in treatment and placebo interventions

Size of study

High risk

n = 21

Maier 2003

Study characteristics

Methods

Randomised, DB, parallel; computer‐generated randomisation

Ff‐up at end of 3 wks

Participants

History of PLP of at least 12 months; PLP of at least 4 on 11‐point numeric rating scale; majority of upper and lower extremity amputations of traumatic aetiology; 36 participants, 29 males; median age in years (range): 62 (28 to 76) in memantine group; 61 (35 to 77) placebo; baseline average pain intensity on 11‐point numeric rating scale (SD): 5.1 (2.13) in memantine group; 5.2 (2.02) placebo

Interventions

  1. memantine at 30 mg/day, once a day, for 3 weeks, oral

  2. placebo

Outcomes

Change in pain intensity on 11‐point numeric rating scale;

Number of participants with > 50% mean pain reduction on 11‐point numeric rating scale;

NNTB for 50% pain reduction (95% CI);

Change in mood/depression score on German validated depression scale;

Change in PDI;

Adverse events;

Dropouts/withdrawals

Notes

Low‐powered study; dosage too low, however this is the limit of clinical tolerability as seen in studies; n = 36

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation

Allocation concealment (selection bias)

Low risk

Doctor not involved in study prepared randomisation; medications prepared in hospital pharmacy

Blinding (performance bias and detection bias)
All outcomes

Low risk

Placebo and memantine (treatment intervention) had the same colour and size (5 mg/capsule)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Last observation carried forward

Selective reporting (reporting bias)

Low risk

All specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes

Other bias

Unclear risk

Baseline characteristics, e.g. time since amputation, dissimilar; longer time period since amputation in the memantine group, but the duration of phantom pain comparable

Size of study

High risk

n = 36

Nikolajsen 1996

Study characteristics

Methods

Controlled clinical trial, DB, cross‐over; 3‐day wash‐out period; non‐involved doctor prepared sealed, numbered envelope for each participant containing order of drugs

Ff‐up at end of infusion: 45 min

Participants

Postamputation stump and phantom pain; upper and lower extremity amputation mostly malignancy in aetiology, rest trauma and infection, reflex dystrophy; 11 participants, 8 male; mean age, yrs (range): 47 (32 to 78); baseline pain intensity on 100‐millimetre VAS: 30.2; median duration of phantom pain, yrs (range): 4 (0.75 to 14)

Interventions

  1. ketamine at 0.5 mg/kg once for 45 minutes, intravenous infusion

  2. placebo (saline)

Outcomes

Change in pain intensity on 0‐to‐100‐millimetre VAS;

Adverse events;

Dropouts/withdrawals;

Other outcomes: McGill Pain Questionnaire, pressure pain threshold, wind‐up‐like pain, thermal stimulus response, temporal summation of heat‐induced pain, reaction time

Notes

Small sample size; n = 11

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

"doctor not involved in study prepared, sealed and numbered envelope for each patient containing order of ketamine and saline administration"

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind design; ketamine and saline same form (IV) and amount; probably done

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants were analysed as to outcomes

Selective reporting (reporting bias)

Unclear risk

Numerical results for pain intensity in VAS not reported but presented in graph as % of baseline values and noted significance; all specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes

Other bias

Low risk

(+) wash‐out period; baseline pain characteristics basically similar in treatment and control periods

Size of study

High risk

n = 11

Robinson 2004

Study characteristics

Methods

Randomised, DB, parallel; randomisation by central pharmacy

Ff‐up at 6 wks

Participants

PLP or residual limb pain; upper and lower limb amputation of vascular, traumatic, cancer, infectious aetiologies; amputation at least 6 months, pain at least 3 months, at least 2 on 0‐to‐10 numerical rating scale; 39 participants, 17 males; mean age, yrs (SD): 44.4 (9.4) in amitriptyline group; 45.3 (13.3) in control; time since amputation, yrs (SD): 11.3 (10.9) in amitriptyline; 10.6 (9.1) control; baseline mean pain intensity on 0‐to‐10 NRS (SD): 3.6 (2.4) amitriptyline; 3.1 (2.6) in control

Interventions

  1. amitriptyline at 10 mg/d titrate each week to max of 125 mg/day for 6 weeks, oral

  2. benztropine mesylate at 0.5 mg/day for 6 weeks, oral

Outcomes

Mean change in pain intensity on 0‐to‐10 NRS;

Change in mood/depression on CES‐D;

Change in function on FIM;

Change in QOL/handicap on CHART;

Adverse events;

Dropouts/withdrawals;

Other outcomes: MPQ, Modified Brief Pain Inventory, satisfaction

Notes

Sample represents only 18% of eligible and may have only selected those refractory to standard treatment; n = 39

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised by pharmacy investigational drug service; probably appropriate random sequence generation

Allocation concealment (selection bias)

Low risk

Random assignment and preparation of medication by hospital pharmacy

Blinding (performance bias and detection bias)
All outcomes

Low risk

"a 7 day supply of medication provided to each participant each week in identical gelatin capsules in plastic holder so that study personnel and participants were blind to medication assignment"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"all pre and post treatments measures were administered by research assistant blinded to the subject assignment"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not all randomised participants were included in analysis (2 (10%) in amitriptyline group excluded)

Selective reporting (reporting bias)

Low risk

All specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes

Other bias

Low risk

Similar baseline characteristics between 2 groups (see characteristics of population above)

Size of study

High risk

n = 39

Schwenkreis 2003

Study characteristics

Methods

Randomised, DB, parallel, computer‐generated randomisation by doctor not involved in study

Ff‐up at end of treatment at 21 days 

Participants

Chronic PLP of at least 12 months; traumatic upper limb amputations; 16 participants, 14 males; median age 62 (35 to 71)

Interventions

  1. memantine titrated up to 30 mg/day x 3 weeks

  2. placebo

Outcomes

Pain intensity;

Dropouts/withdrawals;

Other outcomes: intracortical inhibition; intracortical facilitation

Notes

Small number of participants; n = 16

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation

Allocation concealment (selection bias)

Low risk

Doctor not involved in study performed randomisation; hospital pharmacy prepared medication

Blinding (performance bias and detection bias)
All outcomes

Low risk

"the study medication was produced in hospital pharmacy using capsules of same colour and size for placebo and memantine"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The investigator who undertook the TMS and data analysis was blinded to participants' treatment allocation and assessed pain intensity at same time

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

1 participant in memantine group who did not continue with drug due to adverse events was excluded from study and not included in analysis

Selective reporting (reporting bias)

Low risk

All specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes

Other bias

Low risk

Baseline characteristics between 2 groups similar

Size of study

High risk

n = 16

Smith 2005

Study characteristics

Methods

Randomised, DB, cross‐over; 6 weeks each treatment arm; wash‐out period of 5 weeks; computer‐generated randomisation

Ff‐up at 6 wks

Participants

PLP and residual limb pain, with upper and lower extremity amputations of vascular, traumatic, cancer, infectious aetiology; time since amputation at least 6 months; with average pain intensity of at least 3 on 0‐to‐10 numerical rating scale; 24 participants, 18 males; mean age, yrs (SD): 52.1 (15.5); baseline mean pain intensity on 0‐to‐10 numerical rating scale (SD): 4.38 (2.57)

Interventions

  1. gabapentin titrated from 300 mg to 3600 mg per day for 6 weeks, oral

  2. placebo

Outcomes

Mean change in pain intensity on 0‐to‐10 NRS;

Mean change in mood/depression on CES‐D;

Change in function on FIM;

Change in QOL/handicap on CHART;

Adverse events;

Satisfaction;

Other outcomes: participant rating of global improvement; Modified Brief Pain Inventory; SF‐MPQ sensory score; SF‐MPQ affective score

Notes

Underpowered study; n = 24

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random numbers

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding (performance bias and detection bias)
All outcomes

Low risk

"pharmacy compounded gabapentin and placebo capsules that were identical in appearance so that study investigators and participants could not determine study assignment by the capsules"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Research study nurse contacted each participant to assess pain intensity; probably done

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not all randomised participants were included in final analysis

Selective reporting (reporting bias)

Unclear risk

Adverse events not described in detail/not specified; withdrawals and dropouts not described

Other bias

Low risk

(+) wash‐out period; within‐subject analysis

Size of study

High risk

n = 24

Wiech 2004

Study characteristics

Methods

Randomised, DB, cross‐over, 4 weeks each treatment arm; 2‐week wash‐out phase; randomisation by central pharmacy

Ff‐up at end of treatment at 30 days

Participants

Chronic PLP; all upper extremity amputations of traumatic aetiology; 8 participants, 7 males; mean age in years (SD): 45 (12.51); baseline pain intensity on 0‐to‐100 VAS endpoints (SD): 46.98 (20.38)

Interventions

  1. memantine from 10 mg/day 1st week titrated to 30 mg/day 3rd to 4th week, for 4 weeks oral

  2. placebo

Outcomes

Change in pain intensity on 0‐to‐100 VAS endpoints;

Pain in residual limb;

Adverse events;

Dropouts/withdrawals;

Other outcomes: magnetoencephalography recording

Notes

Small sample size; n = 8

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"the order of treatment was randomised" but not described

Allocation concealment (selection bias)

Low risk

"scientist not involved in study kept a record of treatment assignment"

Blinding (performance bias and detection bias)
All outcomes

Low risk

Member of central pharmacy provided the blinded tablets; placebo substance of identical appearance following same dosage scheme

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants accounted for in analysis

Selective reporting (reporting bias)

Low risk

All specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes

Other bias

Low risk

(+) wash‐out period

Size of study

High risk

n = 8

Wu 2002

Study characteristics

Methods

Randomised, DB, 40 minutes each treatment arm; cross‐over with interval of 24 hours for each infusion; block randomisation

Ff‐up at 30 min after end of infusion

Participants

Persistent postamputation pains > 6 months, lower and upper extremity amputations; 31 participants, 19 males; mean age, yrs (SD): 54 (13); time since amputation in months (SD): 81 (87.4)

Interventions

  1. morphine at 0.2 mg/kg, once given over 40 minutes of intravenous infusion

  2. lidocaine at 4 mg/kg, once given over 40 minutes of intravenous infusion

  3. placebo (diphenhydramine)

Outcomes

Pain intensity on computerised 0‐to‐100 VAS;   

Subjective self reported % pain relief on 0%‐to‐100% numeric scale;

NNTB for 30% PLP pain reduction (95% CI);

Treatment satisfaction scores on 0‐to‐100 numeric scale;

Adverse events;

Dropouts/withdrawals;

Other outcomes: sedation scores

Notes

Study has a power of 80%; carry‐over effects possible, but baseline pain scores in both groups similar as well as short duration of action of drugs; n = 31

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Block randomisation

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding (performance bias and detection bias)
All outcomes

Low risk

All study medications were identical in appearance; investigator administering study medication blinded from intervention; participant and research co‐ordinator blinded

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"during the infusion, the investigator administering the study medication was blinded from the outcome assessment (pain and sedation) and the subject and research coordinators were blinded to the exact timing of study medication administration"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

1 participant dropped out from study due to absence of pain before start of infusion and was not included in analysis

Selective reporting (reporting bias)

Unclear risk

Actual numerical VAS values for pain intensity not reported but presented in graph form and noted significance; all specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes

Other bias

Low risk

Carry‐over effects addressed and discussed; relatively short duration of action of study medications, use of good active placebo, and baseline pain and sedation scores did not differ significantly between 3 days of infusion

Size of study

High risk

n = 31

Wu 2012

Study characteristics

Methods

Randomised, DB, pilot, parallel sequence; 1 group receiving BoNT/A injections and another group receiving combination lidocaine and methylprednisolone injections; “one physician was chosen to implement the treatment protocol for all patients after randomisation.”

Ff‐up at 1, 2, 3 ,4 ,5,6 mos

Participants

Adult lower extremity amputees with PLP or RLP, or both with VAS > 5/10 and unresponsive to conventional treatment

Interventions

  1. Injections of BoNT/A with dosage of 1 mL equal to 50 units of BoNT/A into painful sites; 1 treatment episode

  2. Injections of 1 mL mixture of 0.75 mL of 1% lidocaine and 0.25 mL of lidocaine/methylprednisolone 40 mg/mL into painful sites; 1 treatment episode

Outcomes

Pain intensity by 0‐to‐10 VAS;

Changes in pressure pain tolerance;

Dropouts/withdrawals

Notes

Study has a small size; n = 14

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Technique of allocation not clearly described

Blinding (performance bias and detection bias)
All outcomes

Low risk

Participants were blinded to the type of treatment they received; medications same in appearance, dosage amount, and route

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Evaluators were blinded to the type of treatment participants received

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The study described the dropouts and withdrawals during the course of the study, but unclear whether these were included in the analysis

Selective reporting (reporting bias)

Low risk

All specified outcomes in methods section of published study were reported, although not necessarily as our preferred outcomes

Other bias

Unclear risk

Uncertain if baseline characteristics similar in both groups

Size of study

High risk

n = 14 (but only an initial report)

BI: Barthel Index; BoNT/A: botulinum toxin A; CES‐D: Center for Epidemiologic Studies Depression Scale; CHART: Craig Handicap Assessment and Reporting Technique; CI: confidence interval; DB: double‐blind; FIM: Functional Independence Measure; ff‐up: follow‐up; HADS: Hospital Anxiety and Depression Scale; ICF: intracortical facilitation; ICI: intracortical inhibition; IU: international unit; IV: intravenous; max: maximum; mos: months; MPQ: McGill Pain Questionnaire; n: number of participants; NNTB: number needed to treat for an additional beneficial outcome; NAS: numerical analogue scale; NRS: numerical rating scale; PDI: Pain Disability Index; PLP: phantom limb pain; RLP: residual limb pain; QOL: quality of life; SD: standard deviation; SF‐MPQ: Short‐Form McGill Pain Questionnaire; SIS: Sleep Interference Scale; TMS: transcranial magnetic stimulation; VAS: visual analogue scale; wks: weeks; yrs: years.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Abraham 2002

Sample size of 3

Atesalp 2000

Not reported as randomised or quasi‐randomised; no mention of treatment allocation; no description of double‐blinding

Borghi 2010

Not established PLP/pre‐emptive therapy

Cohen 2011

Not randomised

Elizaga 1994

Not established PLP

Elrazek 2005

Population composed of participants with stump pain; none had PLP

Grant 2008

Pre‐emptive/preventative therapy

Ilfeld 2013

Sample size of 3

Jacobson 1989

Case series

Jacobson 1990

Not established PLP but stump pain

Jaeger 1988

Mixed group of phantom pain and causalgias; no control

Jin 2009

Case series

Karanikolas 2011

Pre‐emptive therapy

Kessel 1987

Non‐randomised, open study

Kukushkin 1996

Pre‐post study; no control group

Licina 2013

Case series

Lirk 2012

Editorial and preventive therapy protocol

Neil 2012

Comment on a study done in 2011

Nikolajsen 1997

Case report

Nikolajsen 2006

Pre‐emptive therapy

Panerai 1990

Mixed diagnoses for central pain

Pinzur 1996

Pre‐emptive therapy

Rogers 1989

Case report

Sato 2008

Case report

Scadding 1982

Mixed diagnoses with no separate analyses for PLP

Van Seventer 2010

Mixed diagnoses with no separate analyses for PLP

Vorobeichik 1997

No description of randomisation, allocation, double‐blinding, who assessed outcomes, withdrawals/dropouts

Wilder‐Smith 2005

Non‐randomised after day 3, where treatment assignment was changed based on response; not all participants were blinded; numerical results for initial responders (first 3 days) not reported

Wu 2008

Included all postamputation pains; did not distinguish PLP from other postamputation pains; no separate analysis for PLP

PLP: phantom limb pain

Characteristics of studies awaiting classification [ordered by study ID]

Buch 2019

Methods

Randomized, double‐blind, placebo‐controlled crossover study

Participants

Amputees with constant postamputation pain. Sample size of 12 (but only 9 were analysed)

Interventions

Peripheral nerve block

Outcomes

The primary outcome was the difference in absolute change between worst pain intensity, either phantom or stump pain, at baseline and at 30 minutes after lidocaine or saline injection.

Notes

Data and analyses

Open in table viewer
Comparison 1. Memantine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Change in pain intensity Show forest plot

2

52

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

0.24 [‐0.31, 0.79]

Analysis 1.1

Comparison 1: Memantine versus placebo, Outcome 1: Change in pain intensity

Comparison 1: Memantine versus placebo, Outcome 1: Change in pain intensity

Open in table viewer
Comparison 2. Gabapentin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Change in pain intensity Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

‐1.16 [‐1.94, ‐0.38]

Analysis 2.1

Comparison 2: Gabapentin versus placebo, Outcome 1: Change in pain intensity

Comparison 2: Gabapentin versus placebo, Outcome 1: Change in pain intensity

Study flow diagram.

Figuras y tablas -
Figure 1

Study flow diagram.

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

Figuras y tablas -
Figure 2

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

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

Figuras y tablas -
Figure 3

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

Comparison 1: Memantine versus placebo, Outcome 1: Change in pain intensity

Figuras y tablas -
Analysis 1.1

Comparison 1: Memantine versus placebo, Outcome 1: Change in pain intensity

Comparison 2: Gabapentin versus placebo, Outcome 1: Change in pain intensity

Figuras y tablas -
Analysis 2.1

Comparison 2: Gabapentin versus placebo, Outcome 1: Change in pain intensity

Table 1. Summary of results

Author, year

Intervention

Treatment

duration

Follow‐up

Outcomes

Results

Overall direction of efficacy

Adverse events

BoNTs

BoNT/A

Wu 2012

  1. BoNT/A, 1 mL = 50 units for each injection site

  2. combi l/m, 1 mL = 0.75 mL of 1% lidocaine and 0.25 mL methylprednisolone 40 mg/mL for each painful site

1 tx episode

At 1, 2, 3, 4, 5, 6 mos

Change in VAS, change in pressure pain tolerance

No significant change in phantom pain and pressure pain tolerance

Not described

NMDA antagonists

Memantine

Maier 2003

  1. memantine 30 mg/d; oral

  2. placebo

3 weeks

At end of

3 weeks

Pain intensity 11‐point NRS; number of participants with > 50% pain reduction; NNTB;

mood; disability; adverse events

No sig diff in change in pain level, in number of

participants with > 50% pain relief; depression scores; disability indices

in 2 grps; overall number severe events higher in

memantine grp

Vertigo,

tiredness,

headache,

nausea,

restlessness,

excitation,

cramps

Wiech 2004

  1. memantine titrated up to 30 mg/d; oral

  2. placebo

4 weeks

each

treatment

arm

At end

of 4

weeks

of each

arm

Pain intensity 0‐to‐100 VAS; MEG recording; adverse events

No sig diff in change in pain

intensity, cortical

reorganisation in both grps

Nausea,

fatigue,

dizziness,

agitation,

headaches

Schwenkreis 2003

  1. memantine titrated up to 30 mg/d; oral

  2. placebo

3 weeks

At end of

3 weeks

Pain intensity 11‐point NRS; ICI; ICF

No sig diff in pain intensity;

enhanced ICI; reduced ICF

Not

described

Dextromethorphan

Abraham 2003

  1. dextromethorphan 120 mg/d; oral

  2. dextromethorphan 180 mg/d; oral

  3. placebo

10 days

each

treatment

arm 

At end of

10 days of

each arm

Number of participants with ≥ 50% pain relief; feeling of well‐being; sedation score; adverse events

Dextromethorphan grps with ≥ 50% pain relief; with sig better feeling of well‐being scores; with sig lower sedation scores

+

None

reported

Ketamine

Nikolajsen 1996

  1. ketamine 0.5 mg/kg once, IV infusion

  2. placebo

45 min

each

treatment

arm

At end of IV infusion

Pain intensity 0‐to‐100‐millimetre VAS; adverse events; McGill; pressure pain threshold; wind‐up like pain; thermal stimulus response; temporal

summation of heat‐induced pain; reaction time

Sig dec in pain intensity; in

pain evoked by mechanical

stimulation; inc in pressure

pain threshold; no alteration in temperature sensitivity in

ketamine group

+

Insobriety,

discomfort,

elevation of

mood

Eichenberger 2008

  1. ketamine 0.4 mg/kg once, IV infusion

  2. calcitonin 200 IU, once, IV infusion

  3. combination ketamine/calcitonin, IV

  4. placebo

1 hour

each

arm

At 30, 60

mins, 48

hours after

infusion

Pain intensity; number of participants with ≥ 50% pain reduction on 10‐centimetre VAS; basal sensory assessment; adverse events

Sig dec pain intensity in

ketamine alone and combination vs placebo and calcitonin; sig inc in number of responders in ketamine alone and combination vs placebo and calcitonin; sig

inc in electrical thresholds with

combination treatment but no

change in pressure or heat thresholds

+

Loss of

conscious

ness, light

sedation,

light visual

hallucination,

hearing

impairment,

position/

feeling

impairment

Anticonvulsants

Gabapentin

Bone 2002

  1. gabapentin titrated up to 2400 mg or max tolerable dose; oral

  2. placebo

6 weeks

each arm

Weekly

and at

end of

6 weeks

Pain intensity 100‐millimetre VAS; pain intensity

difference; depression

score (HADS); function (BI); sleep (SIS); no. of rescue tabs; adverse events

Significantly greater pain

intensity diff with gabapentin at end of treatment; no sig diff in depression score, function,

sleep, no. of rescue tablets with the treatments

+a

b

Somnolence, dizziness,

headache,

nausea

Smith 2005

  1. gabapentin titrated up to 3600 mg/d; oral

  2. placebo

6 weeks

At end of 6 weeks of each

arm

Pain intensity 0‐to‐10 NRS;

depression score (CES‐D); function (FIM);

handicap (CHART);

satisfaction; global

improvement rating; pain

inventory; McGill

No sig group diff on any outcomes at end of treatment

c

Not

described

Antidepressants

Amitriptyline

Robinson 2004

  1. amitriptyline 10 mg/d titrated to max of 125 mg/d; oral

  2. benztropine mesylate 0.5 mg/d; oral

6 weeks

At end of 6 weeks

Pain intensity 0‐to‐10 NRS;

depression score (CES‐D); function (FIM); handicap (CHART); pain inventory; McGill; satisfaction

No sig group diff on any outcomes at end of treatment

Dry mouth

(more severe),

dizziness

Calcitonins

Jaeger 1992

  1. s‐calcitonin 200 IU, IV infusion

  2. saline

20‐minute

IV infusion;

once

24 hours

after

infusion

(DB);

7 to 152

days,

weekly

(open

phase)

Pain intensity 0‐to‐10 NAS in open phase/long term; number of participants with > 50%, 75% pain relief; adverse events

Sig dec in median pain intensity with s‐calcitonin at 24

hours after infusion; at 1 yr,

62% of participants with 75% pain

reduction

+

Headache,

vertigo,

nausea,

vomiting,

phantom

sensation,

drowsiness,

hot/cold

flushes

Eichenberger 2008

  1. ketamine 0.4 mg/kg, once, IV infusion

  2. calcitonin 200 IU, once, IV infusion

  3. combination ketamine/calcitonin, IV

  4. placebo

1 hour

each arm

At 30, 60

mins, 48

hours after

infusion

Pain intensity; number of

participants with ≥ 50% pain relief on 10‐centimetre VAS; basal sensory assessments; adverse effects

No sig dec in pain intensity with calcitonin vs placebo at 48 hrs; number of responders

not significantly different from

placebo

Drowsiness,

nausea,

facial

flushing,

hot/cold

flushes,

dizziness

Opioids

Morphine

Huse 2001

  1. Morphine sulfate titrated up to 300 mg/d or max tolerable dose; oral

  2. placebo

4 weeks

each arm

(DB)

End of each

treatment

phase of 4

weeks

Pain intensity 10‐centimetre VAS; number of participants with > 50% pain reduction; depression score; pain‐related self assessment scale;

WHYMPI; BSS; psycho‐

physical thresholds;

2‐point discrimination;

attentional performance;

MEG

Sig pain reduction during morphine; 42% with > 50% pain relief; 8% with 25% to 50% pain relief during morphine; no sig change in perception and

pain thresholds; significantly

lower attentional performance

during morphine; scores on pain experience scale, depression score, WHYMPI, BSS with no sig relationship with pain reduction; 2 of 3 with clear cortical reorganisation

+

Constipation only sig

adverse

effect among

others, e.g.

tiredness,

dizziness,

sweating,

micturition

difficulty,

vertigo,

itching,

respiration

Wu 2002

  1. morphine 0.2 mg/kg, IV infusion

  2. lidocaine 4 mg/kg, IV infusion

  3. placebo (diphenhydramine)

40 mins of IV

infusion

30 mins

after end of infusion

Pain relief 0‐to‐100% numeric scale; NNTB for

30% pain reduction;

satisfaction; sedation

scores; adverse events

Sig dec in phantom and stump pain intensity during IV morphine; NNTB 1.9 (95% CI 1.3 to 3.7); significantly higher satisfaction with morphine; no sig diff in sedation scores

+

Sedation

(but no sig

diff with other groups)

Local anaesthetics

Lidocaine

Wu 2002

  1. morphine 0.2 mg/kg, IV infusion

  2. lidocaine 4 mg/kg, IV infusion

  3. placebo (diphenhydramine)

40 mins

of IV

infusion

30 mins

after end of

infusion

Pain relief 0‐to‐100% numeric scale; NNTB for

30% pain reduction;

satisfaction; sedation

scores; adverse events

No sig dec in PLP vs placebo; NNTB 3.8 (95% CI 1.9 to 16.6); significantly higher satisfaction with lidocaine vs

placebo; no sig diff in sedation scores

Sedation

scores not

significantly

different

from

placebo

Bupivacaine

Casale 2009

  1. bupivacaine 2.5 mg/mL, 1mL, contralateral myofascial injection

  2. placebo (saline)

Injections

given

once

After 1

hour

Pain intensity 0‐to‐10 VAS from 0 no pain to 10 worst pain ever experienced;

pain intensity difference;

phantom sensation;

mirror displacement

in healthy limbs; adverse

effects

Sig pain relief with bupivacaine; reduction in phantom sensation in 6 of 8 participants

+

 None

BI, Barthel Index; BoNT/A, botulinum toxin A; BSS, Brief Stress Scale; CES‐D, Center for Epidemiologic Studies Depression Scale; CHART, Craig Handicap Assessment and Reporting Technique; CI, confidence interval; combo, combination; d, day; DB, double‐blind; dec, decrease; diff, difference; dx, diagnosis; FIM, Functional Independence Measure; grp, group; grps, groups; HADS, Hospital Anxiety and Depression Scale; ICI, intracortical inhibition; ICF, intracortical facilitation; inc, increase; IU, international units; IV, intravenous; l/m, lidocaine/methylprednisolone; max, maximum; MEG, magnetoencephalography; min, minutes; mos, months; NAS, numerical analogue scale; NNTB, number needed to treat for an additional beneficial outcome; NRS, numerical rating scale; PLP, phantom limb pain; sig, significant; SIS, Sleep Interference Scale; tx, treatment; VAS, visual analogue scale; WHYMPI, West Haven‐Yale Multidimensional Pain Inventory; yr, year; apain intensity; bmood, sleep, function; cpain intensity, mood, function, handicap, satisfaction

Figuras y tablas -
Table 1. Summary of results
Comparison 1. Memantine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Change in pain intensity Show forest plot

2

52

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

0.24 [‐0.31, 0.79]

Figuras y tablas -
Comparison 1. Memantine versus placebo
Comparison 2. Gabapentin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Change in pain intensity Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

‐1.16 [‐1.94, ‐0.38]

Figuras y tablas -
Comparison 2. Gabapentin versus placebo