Scolaris Content Display Scolaris Content Display

Intervenciones farmacológicas para el tratamiento del dolor del miembro fantasma

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

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.

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.

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.

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.

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.

Jaeger 1992 {published data only}

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

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.

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.

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.

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.

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.

Wieck 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.

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.

Referencias de los estudios excluidos de esta revisión

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. The Clinical Journal of Pain 2002;18(5):282‐5.

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.

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 and Analgesia 2010;111(5):1308‐15.

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.

Elizaga 1994 {published data only}

Elizaga AM, Smith DG, Sharar SR, Edwards WT, Hansen ST. 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.

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.

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.

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.

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.

Jaeger 1988 {published data only}

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

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.

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.

Kessel 1987 {published data only}

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

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] [Russian]. Anesteziologiia i Reanimatologiia 1996;4:39‐42.

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.

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.

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.

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. The Journal of Bone and Joint Surgery. American volume 1996;78(10):1501‐5.

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.

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.

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.

Vorobeichik 1997 {published data only}

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

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.

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.

Referencias adicionales

Azria 2002

Azria M. Possible mechanisms of the analgesic action of calcitonin. Bone 2002;30(5):80S‐83S.

Bileviciute‐Ljungara 2001

Bilevicuite‐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. The Journal of Neuroscience 1997;17(14):5503‐8.

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.

Eisenberg 2009

Eisenberg E, McNicol ED, Carr DB. Opioids for neuropathic pain. Cochrane Database of Systematic Reviews 2006, Issue 3. [DOI: 10.1002/14651858.CD006146]

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. Neuro Report 1994;5(18):2593‐7.

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.

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.

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.

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.

Hayes 2004

Hayes C, Armstrong‐Brown A, Burstal R. Perioperative intravenous ketamine infusion for the prevention of persistent post‐amputation pain:a randomized, controlled trial. Anaesthesia and Intensive Care 2004;32(3):330‐8.

Higgins 2011

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

Jadad 1996

Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJM, Gavaghan DJ, et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary?. Controlled Clinical Trials 1996;17:1‐12.

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.

Khan 1996

Khan KS, Daya S, Jadad A. The importance of quality of primary studies in producing unbiased systematic reviews. Archives of Internal Medicine 1996;156(6):661‐6.

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.

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.

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 2011

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

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.

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.

Ramachandran 1992

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

Saarto 2007

Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database of Systematic Reviews 2007, Issue 4. [DOI: 10.1002/14651858.CD005454.pub2]

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.

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.

Van Tulder 2003

Van Tulder M, Furlan A, Bombardier C, Bouter L, Editorial Board of the Cochrane Collaboration Back Review Group. Updated method guidelines for systematic reviews in the Cochrane Collaboration Back Review Group. Spine 2003;28(12):1290‐9.

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.

Wilson 2008

Wilson J, Nimmo A, Fleetwood‐Walker S, Colvin L. A randomised double blind trial of the effect of pre‐emptive epidural ketamine on persistent pain after lower limb amputation. Pain 2008;135(1‐2):108‐18.

Wolff 2011

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

Ypsilantis 2010

Ypsilantis E, Tang T. Pre‐emptive analgesia for chronic limb pain after amputation for peripheral vascular disease: a systematic review. Annals of Vascular Surgery 2010;24(8):1139‐46.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Abraham 2003

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 

Participants

Severe phantom pain for at least one 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 patients 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; drop‐outs/withdrawals

Results: 1) & 2) all patients reported ≥ 50% pain relief; Dextromethorphan treatments with significantly better scores for feelings of well‐being at P = 0.025; Sedation significantly lower in dextromethorphan groups P = 0.01; no side effects recorded throughout drug treatment; Drop‐outs/withdrawals: None

Notes

Limitations related to dosing and small sample size

QS: Jadad score 3; Van Tulder score 7

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

Other bias

High risk

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

Bone 2002

Methods

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

Participants

Patients with phantom pain ? 6 months with pain intensity ?40 mm on 100 mm 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) : treatment group 6.1 (1.8); placebo 6.7 (1.9)

Interventions

1) gabapentin titrated in increments of 300 mg up to 2, 400 mg or maximal tolerable dose for 6 weeks; oral

2) placebo

Outcomes

Change in pain intensity 100 mm VAS at end of treatment week 6 vs. baseline

1) pre 6.1 (1.8); post 2.9 (2.2)

2) pre 6.7 (1.9); post 5.1 (2.2)

Mean Pain intensity difference at end of treatment week 6 (SD)

1) 3.2 (2.1)

2) 1.6 (0.7)

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

1) pre 1.5 (0.9); post 1.45 (1)

2) pre 1.8 (0.9); post 1.6 (1.2)

Median change in mood on HADS (range)

1) pre 14 (5 to 25); post 12 (4 to 23)

2) pre 15 (5 to 25); post 14 (5 to 25)

Median change in function on BI (range)

1) pre 90 (70 to 105); post 85 (70 to 105)

2) pre 85 (65 to 100); post 87 (65 to 105)

Median change in sleep on SIS (range)

1) pre 4 (2 to 5); post 3 (1 to 5)

2) pre 4 (2 to 5); post 4 (1 to 5)

Other outcomes: number of rescue tablets

Adverse events: somnolence, dizziness, headache, nausea

Drop‐outs/withdrawals: 

1) 2 : one did not complete and another one protocol violation

2) 3 : two did not complete and one withdrew consent

Results: Significant mean pain intensity differences between two groups at 6th week in favour of gabapentin; no significant differences in rest of outcomes

Notes

For lifestyle indices, sample size may be too small to rule out type 2 error; used between group analysis for comparisons
QS: Jadad score 5; Van Tulder score 9

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

it is likely that the expected outcomes were reported

Other bias

Low risk

carry‐over effect in cross‐over trial but probably low risk because baseline VAS pain score before placebo/gabapentin are not significantly different

Casale 2009

Methods

Randomised, DB, cross‐over with 72 hours wash‐off period; Computer‐generated randomisation

Participants

Patients with PLP of at least 6 months, lower extremity amputation of traumatic and vascular in aetiology; 8 participants, 6 males; mean age, yrs (SD): 70.1 (7.7); Baseline pain intensity on 0‐10 VAS, mean (SD): 7.9 (0.8) in 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 0 to 10 VAS, (SD)

1) pre 7.9 (0.8); post 2.6 (1.2)

2) pre 7.6 (0.7); post 6.1 (1.6)

Mean difference pain intensity (SD)

1) ‐5.3 (1.4)

2) ‐1.5 (1.3)

Adverse effects

Drop‐outs/withdrawals

Other outcomes: phantom sensation, mirror displacement in healthy limbs

Results: Significant pain relief with bupivacaine contralateral myofascial injection P = 0.003. Adverse effects: no clinical signs of cardiovascular or respiratory side effects; no subjective reactions reported; no stinging sensation reported; Drop‐outs/withdrawals: None

Notes

small number of patients; preliminary results

QS: Jadad score 4; Van Tulder score 10

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"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

all patients accounted for

Selective reporting (reporting bias)

Low risk

it is likely that the expected outcomes were reported

Other bias

Low risk

carry‐over effect in cross‐over, but probably low risk as baseline pain intensity during anaesthetic and saline not significantly different

Eichenberger 2008

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

Participants

Patients with 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 cm VAS scale; 20 participants; 15 males; Median age, yrs (range): 57 (19.3 to 72.7); Mean baseline pain intensity on 10 cm VAS: 4.32; Duration of phantom pain, yrs: 12.41

Interventions

1) ketamine at 0.4 mg/kg, once, one hour intravenous infusion

2) calcitonin  at 200 IU once, one hour intravenous infusion

3) combination ketamine / calcitonin at 200 IE calcitonin and 0.4 mg/kg ketamine once, one hour intravenous infusion

4) placebo (saline)

Outcomes

Number of patients with > 50% pain reduction on 10 cm VAS

1) 6 out of 10

2) 2 out of 20

3) 12 out of 20

4) 1 out of 19

Change in pain intensity on 10 cm VAS

Adverse effects:

Drop‐outs/withdrawals

Other outcomes: Basal Sensory assessments

Results: Ketamine alone and its combination with calcitonin but not calcitonin decreased pain intensity during and after administration of drugs compared to placebo; combination calcitonin and ketamine not superior to ketamine alone; adverse effects with 1) ketamine: severe loss of consciousness; others: light sedation and light visual hallucination/hearing impairment/impairment of position/feeling;
2) calcitonin: facial flushing, nausea, light sedation, dizziness; 3) combination ketamine/calcitonin: facial flushing, nausea, light sedation, dizziness, light visual hallucination, hearing impairment, position/feeling impairment; Drop‐outs/withdrawals: One from the ketamine alone group did not receive the saline infusion. Two from the placebo group did not get the heat and electrical stimulation

Notes

relatively small sample size; wide range duration phantom pain; ketamine alone was given to only 10 participants

QS:Jadad score 5; Van Tulder score 9

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 one group but not related to outcome

Selective reporting (reporting bias)

Unclear risk

added methods (introduction of fourth session)

Other bias

Unclear risk

carry‐over effect

Huse 2001

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 patients randomised and kept code

Participants

Patients with phantom pain, with upper and lower extremity amputations, at least 3 in 10 cm visual analog scale; 12 participants, 10 males; mean age, yrs (SD): 50.58 (14.01); Mean baseline pain intensity on 10 cm 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 / max tolerable dose for 4 weeks, oral

2) placebo

Outcomes

Change in pain intensity on 0 to 10 cm VAS (SD)

1) pre 4.65 (1.06); post 3.26 (1.59)

2) pre 4.65 (1.06); post 3.99 (1.23)

Number of patients with pain reduction of > 50% (10 cm VAS)

1) 5 out of 12

2) 1 out of 12

Change in mood /depression on Self‐rating Depression Score

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

Other outcomes: Pain‐related self‐assessment scale; active coping and catastrophizing using West‐Haven Yale Multidimensional Pain Inventory; Brief stress scale; psychophysical thresholds, 2‐point discrimination; attentional performance with d‐2 test, magnetoenkephalography

Results: significantly lower pain intensity during morphine sulphate compared to the baseline (P = 0.01) and significantly lower compared to the placebo phase (P = 0.036); Significant attentional deficits with morphine sulfate (MST); Adverse effects: tiredness, dizziness, sweating, constipation*, micturition diff, nausea, vertigo, itching, short of respiration. Side effects were moderate and higher for morphine vs. placebo. Significance noted only in constipation; drop‐outs/withdrawals: None during DB phase; mood/depression, coping & catastrophizing, stress and the other psychosocial variables with no significant relationship to pain reduction

Notes

Small sample size; cortical reorganization results based on 3 participants (open phase)

QS: Jadad score 4; Van Tulder score 8

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 patients randomised and kept code

Blinding (performance bias and detection bias)
All outcomes

High risk

"the participants as well as treating physicians were able to identify the morphine treatment but not the placebo treatment'; blinding may have been broken

Blinding of outcome assessment (detection bias)
All outcomes

High risk

it is likely that blinding could have been broken

Incomplete outcome data (attrition bias)
All outcomes

Low risk

all patients accounted for in analysis of outcomes

Selective reporting (reporting bias)

Low risk

it is likely that the expected outcomes were reported

Other bias

Low risk

carry‐over effect addressed

Jaeger 1992

Methods

Controlled clinical trial, cross‐over; 2‐hours 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

Participants

Patients with 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 analog scale; 21 participants, 12 males; Median age yrs (range): 59 (20 to 78)

Interventions

1) s‐calcitonin at 200 IU, once, 20 minutes intravenous infusion 

2) saline

Outcomes

Change in pain intensity on 0 to 10 numeric analog scale, median

1) pre 7; post 4

2) pre 7; post 7

Number of patients with pain reduction of > 50%

Long‐term (at 1 yr): number of patients with reduction of > 75%

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

Adverse events

Drop‐outs/withdrawals

Results: significant change in pain intensity in calcitonin phase; 19/21 with pain reduction > 50%; 8/13 surviving patients with pain reduction > 75% at 1 yr; Adverse events: headache, vertigo, nausea, vomiting, augmentation of phantom sensation, drowsiness, hot/cold flushes; Drop‐outs/withdrawals: In the long‐term follow‐up 6 months to 2 years, a total of 8 participants have succumbed

Notes

QS: Jadad score 3; Van Tulder score 6

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 patients 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

Other bias

Unclear risk

carry‐over effects in cross‐over design

Maier 2003

Methods

Randomised, DB, parallel; computer‐generated randomisation

Participants

Patients with history of at least 12 months phantom pain of at least 4 on 11‐point numeric rating scale; upper and lower extremity amputations of traumatic aetiology in majority; 36 patients; 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, mean (SD)

1) pre 5.1 (2.1); post 3.8 (2.3)

2) pre 5.1 (2); post 3.2 (1.6)

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

1) 10/18

2) 6/18

NNT for 50% pain reduction (95%CI): 4.5 (95%CI 2.1 to 10.6)

Mean change in mood / depression score on German validated depression scale (SD)

1) 22 (12)

2) 31 (18)

Mean change in PDI (SD)

1) 22.1 (12)

2) 17 (7)

Adverse events: vertigo, tiredness, headache, nausea, restlessness, excitation, cramps

Drop‐outs/withdrawals:

1) two patients due to side effects

2) three patients due to insufficient analgesia

Results: No significant difference in change in pain level in two groups; Mean pain relief was similar in both groups; no significant change in depression score; Over‐all number in severe events higher in memantine group (P < 0.05)

Notes

Low powered study; Dosage too low, however, this is the limit of clinical tolerability as seen in studies

QS: Jadad score 5; Van Tulder score 9

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 verum had same colour and size"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

last observation carried forward

Selective reporting (reporting bias)

Low risk

it is likely that all expected outcomes were reported

Other bias

Unclear risk

baseline characteristics e.g. time since amputation, not similar; Longer time period since amputation in the memantine group but the duration of phantom pain comparable

Nikolajsen 1996

Methods

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

Participants

Patients with 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 mm 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 mm VAS

Adverse events

Drop‐outs/withdrawals

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

Results: All patients with significant decrease in pain intensity in ketamine group vs placebo P = 0.05; Adverse events insobriety, discomfort, elevation of mood; Drop‐outs/withdrawals: None

Notes

small sample size

QS: Jadad score 3; Van Tulder score 8

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 patients 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

Other bias

Unclear risk

carry‐over effects in cross‐over design

Robinson 2004

Methods

Randomised, DB, parallel; randomisation by central pharmacy

Participants

Patients with 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 patients, 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/d, daily for 6 weeks, oral

2) benztropine mesylate at 0.5 mg/d, daily for 6 weeks, oral

Outcomes

Mean change in pain intensity on 0 to 10 NRS (SD)

1) pre 3.6 (2.4); post 3.1 (2.7)

2) pre 3.1 (2.6); post 3.1 (2.9)

Change in mood / depression on CES‐D (SD)

1) pre 16.2 (11.9); post 12.9 (8.5)

2) pre 16.4 (12.4); post 1.4 (13.1)

Change in function on FIM (SD)

1) pre 78.4 (3.8); post 74.5 (18.8)

2) pre 78.3 (4.2); post 79.1 (3.3)

Change in QOL/handicap on CHART

1) pre 417 (76); post 360 (142)

2) pre 422 (82); post 417 (75)

Other outcomes: MPQ, Modified Brief Pain Inventory, satisfaction

Adverse effects

Drop‐outs/withdrawals

Results: No significant change in pain intensity, depression scores, function, QOL; Adverse effects: dry mouth*, drowsiness, tiredness/fatigue, BOV, constipation dizziness, heartburn, poor sleep, palpitation, nausea, better sleep, urine retention, diarrhoea, tinnitus, tremor, sweating , headaches; Drop‐outs/withdrawals: two due to side effects

Notes

sample represents only 18% of eligible and may have only selected those refractory to standard treatment;

QS Jadad score 5; Van Tulder score 9

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 (two (10%) in amitriptyline group excluded)

Selective reporting (reporting bias)

Low risk

it is likely to have included the expected outcomes

Other bias

Low risk

similar baseline characteristics between 2 groups

Schwenkreis 2003

Methods

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

Participants

Patients with 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; intracortical inhibition;  intracortical facilitation

Results: no significant difference in change of pain intensity in both groups; significantly increased ICI in memantine group; significantly reduced ICF in memantine group; no correlation between changes in pain intensity and neurophysiological parameters

Notes

1 participant in memantine group discontinued in study, excluded from analysis; small number of participants

QS: Jadad score 5; Van Tulder 9

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 did 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 patients' treatment allocation and assessed pain intensity at same time

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

one patient in memantine who did not continue with drug due to adverse effects was excluded from study and not included in analysis

Selective reporting (reporting bias)

Low risk

likely to have included the expected outcomes

Other bias

Low risk

baseline characteristics between two groups similar

Smith 2005

Methods

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

Participants

Patients with 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 3,600 mg per day for 6 weeks, oral

2) placebo

Outcomes

Mean change in pain intensity on 0 to 10 NRS (SD)

1) pre 4.38 (257); post 3.43 (2.45)

2) pre 4.09 (2.44); post 3.6 (2.67)

Mean change in mood/depression on CES‐D (SD)

1) pre 17.5 (10.71); post 3.74 (10.17)

2) pre 18.58 (12.67); post 14.81 (9.82)

Change in function on FIM

Change in QOL / handicap on CHART

Adverse effects

Satisfaction

Other outcomes: Participant rating of global improvement; modified brief pain inventory; SF MPQ sensory score; SF MPQ affective score

Results: No significant difference in change in pain intensity, mood/depression scores, function, QOL, satisfaction; Adverse effects: not described but benefit of drug outweighed side effects compared with one third of placebo who reported benefit; Drop‐outs/withdrawals: Not described in paper

Notes

underpowered study

QS: Jadad score 4; Van Tulder score 7

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 sot 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 patients were included in final analysis

Selective reporting (reporting bias)

Low risk

it is likely that the expected outcomes were reported

Other bias

Low risk

carry‐over effects addressed, within‐subject analysis

Wieck 2004

Methods

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

Participants

Patients with chronic PLP; all upper extremity amputations of traumatic aetiology; eight participants; seven 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‐4th week, for 4 weeks oral

2) placebo

Outcomes

Change in pain intensity on 0 to 100 VAS endpoints

1) pre 46.98 (20.38); post 51.51 (20.61)

2) pre 46.98 (20.38); post 49.46 (21.11)

Magnetoenkephalographic recording

Pain in residual limb

Adverse events

Drop‐outs/withdrawals

Results: No significant differences in change in pain intensity in both groups; No significant differences in cortical reorganization; no significant difference in residual limb pain; adverse events: nausea, fatigue, dizziness, agitation, headaches; Drop‐outs/withdrawals: None

Notes

small sample size

QS: Jadad score 3; Van Tulder score 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 scheme of dosage

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

all patients accounted for in analysis

Selective reporting (reporting bias)

Low risk

it is likely that the expected outcomes reported

Other bias

Low risk

carry‐over effects addressed

Wu 2002

Methods

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

Participants

Patients with 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, mean (SD)

1) 47.9 (38.2)

2) 25.8 (31.2)

3) 3.2 (10.1)

NNT for 30% PLP pain reduction (95%CI)

1) 1.9  (95% CI 1.3 to 3.7)

2) 3.8 (95% CI:1.9 to 16.6)

Other outcomes: treatment satisfaction scores on 0 to 100 numeric scale, sedation scores

Adverse events

Drop‐outs/ withdrawals

Results: Morphine significantly decreases both stump (P < 0.01) and phantom pain (P < 0.001) vs. placebo; Lidocaine significantly decreases stump pain vs. placebo (P < 0.01) but not phantom pain (P > 0.05); Adverse events: sedation with morphine but no significant difference vs. other groups; Drop‐outs /withdrawals: 1 dropped out because of absence of pain before start of infusions

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

QS: Jadad score 5; Van Tulder score 9

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; subject and research coordinator 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

One 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

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

BI: Barthel Index; CES‐D: Center Epidemiologic Depression Scale; CHART: Craig Handicap Assessment and Reporting Technique; CI: confidence interval; DB: double blind; FIM: Functional Independence Measure; HADS: Hospital Anxiety and Depression Score; ICF: intracortical facilitation; ICI: intracortical inhibition; MPQ: McGill Pain Questionnaire; NNT: number needed to treat; NRS: numerical rating scale; NRS: numerical rating scale; PDI: Pain Disability Index; PLP: phantom limb pain; QOL: quality of life; QS: quality score; SD: standard deviation; SIS: Sleep Interference Scale; TMS, transcranial magnetic stimulation; VAS: visual analog scale; yrs: years

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Abraham 2002

Sample size of three

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; interventional

Elizaga 1994

Not established PLP

Elrazek 2005

Population composed of patients with stump pain; none had PLP

Grant 2008

Pre‐emptive/preventative therapy

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 and post study; no control group

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 phantom pain

Vorobeichik 1997

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

Wilder‐Smith 2005

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

Wu 2008

Included all postamputation pains; phantom pain was not distinguished from other postamputation pains; no separate analysis for phantom pain

PLP: phantom limb pain

Data and analyses

Open in table viewer
Comparison 1. memantine vs. placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

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 vs. placebo, Outcome 1 change in pain intensity.

Comparison 1 memantine vs. placebo, Outcome 1 change in pain intensity.

Open in table viewer
Comparison 2. gabapentin vs. placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 change in pain intensity Show forest plot

2

Mean Difference (Fixed, 95% CI)

‐1.16 [‐1.94, ‐0.38]

Analysis 2.1

Comparison 2 gabapentin vs. placebo, Outcome 1 change in pain intensity.

Comparison 2 gabapentin vs. placebo, Outcome 1 change in pain intensity.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 1

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 2

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

Comparison 1 memantine vs. placebo, Outcome 1 change in pain intensity.
Figuras y tablas -
Analysis 1.1

Comparison 1 memantine vs. placebo, Outcome 1 change in pain intensity.

Comparison 2 gabapentin vs. placebo, Outcome 1 change in pain intensity.
Figuras y tablas -
Analysis 2.1

Comparison 2 gabapentin vs. placebo, Outcome 1 change in pain intensity.

Table 3. Jadad 1996 Criteria

Author, Year

1a.

1b.

 

1c.

2a.

 

2b.

2c.

3.

 

Total Score

Abraham 2003

0

0

0

1

1

0

1

3 / 5

Bone 2002

1

1

0

1

1

0

1

5 / 5

Casale 2009

1

1

0

1

1

0

0

4 / 5

Eichenberger 2008

1

1

0

1

1

0

1

5 / 5

Huse 2001

1

0

0

1

1

0

1

4 / 5

Jaeger 1992

1

0

0

1

0

0

1

3 / 5

Maier 2003

1

1

0

1

1

0

1

5 / 5

Nikolajsen 1996

1

0

0

1

1

0

0

3 / 5

Robinson 2004

1

1

0

1

1

0

1

5 / 5

Schwenkreis 2003

1

1

0

1

1

0

1

5 / 5

Smith 2005

1

1

0

1

1

0

0

4 / 5

Wieck 2004

1

0

0

1

1

0

0

3 / 5

Wu 2002

1

1

0

1

1

0

1

5 / 5

Figuras y tablas -
Table 3. Jadad 1996 Criteria
Table 4. Van Tulder 2003 Criteria

Author, Year

A

B

C

D

E

F

G

H

I

J

K

Total Score

Abraham 2003

0

1

1

1

1

0

0

0

1

1

1

7 / 11

Bone 2002

1

1

1

1

1

0

0

1

1

1

1

9 / 11

Casale 2009

1

1

1

1

1

1

0

1

1

1

1

10 / 11

Eichenberger 2008

1

1

1

1

1

0

0

1

1

1

1

9 / 11

Huse 2001

0

1

1

1

1

0

1

0

1

1

1

8 / 11

Jaeger 1992

0

0

1

1

0

0

0

1

1

1

1

6 / 11

Maier 2003

1

1

1

1

1

0

0

1

1

1

1

9 / 11

Nikolajsen 1996

0

1

1

1

1

0

0

1

1

1

1

8 / 11

Robinson 2004

1

1

1

1

1

1

1

0

1

1

0

9 / 11

Schwenkreis 2003

1

1

1

1

1

1

0

1

1

1

0

9 / 11

Smith 2005

1

1

1

1

1

1

0

0

0

1

0

7 / 11

Wieck 2004

1

1

1

1

1

0

0

0

1

1

1

8 / 11

Wu 2002

1

1

1

1

1

0

1

1

1

1

0

9 / 11

Figuras y tablas -
Table 4. Van Tulder 2003 Criteria
Table 1. Summary of Characteristics of Included Studies

Author, yr

Number of patients

Characteristics of population

Methodology

Follow‐up

Drop‐outs/ withdrawals

Quality score

 

 

 

 

 

 

 

Jadad

Van Tulder

NMDA antagonists

 

 

 

 

 

 

 

Memantine

 

 

 

 

 

 

 

Maier 2003

36

 

29 males, median age 62 yrs; majority traumatic upper and lower limb amputation; at least 12 mos PLP history;  at least 4 on 11pt NAS

Randomised, DB, parallel; CGR

 

End of 3 wks

 

Two in treatment group due to side affects; three in placebo due to insufficient analgesia

5

9

Wieck 2004

 

8

 

7 males, mean age 45 yrs; chronic PLP; traumatic upper limb amputations

 

Randomised, DB, cross‐over; 2 wk wash‐out; central pharmacy randomisation

End of treatment at 30 days

 

None

 

3

8

Schwenkreis 2003

 

16

 

14 males; median age 62 yrs; at least 12 mos PLP; upper limb amputation

Randomised, DB, parallel, CGR

 

End of treatment at 21 days 

One in treatment group due to side effects

5

9

Dextromethorphan

 

 

 

 

 

 

 

 Abraham 2003

 10

5 males, mean age 50 yrs; average of  4.8 mos PLP; severe pain at least 1 mo; majority upper, lower limb malignant amputations; others vascular and trauma

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

After 10 days of each treatment period (DB phase)

None 

 3

 7

 Ketamine

 

 

 

 

 

 

 

 Nikolajsen 1996

 11

 8 males; mean age 47 yrs; average of 4 yrs PLP; with postamputation and phantom pain; upper, lower limb amputation; mostly malignant others trauma, infection, reflex sympathetic dystrophy

 Randomised, DB, cross‐over, 3 days wash‐out; non‐involved doctor prepared, sealed, numbered envelope for each patient containing order of drug

At end of infusion: 45 min

None

 3

 8

 Eichenberger 2008

 20

15 males, median age 57 yrs; mean pain intensity 4.32 on 10 cm VAS, average of 12 yrs PLP

 Randomised, DB, cross‐over, time between infusions 48 hours; drawing of lots by person not involved in study

 30,60 min, 48 hrs after infusion

 One from ketamine alone group did not get saline; two from placebo did not get heat, electrical stimulation

 5

 9

  Anticonvulsants

 

 

 

 

 

 

 

 Gabapentin

 

 

 

 

 

 

 

 Bone 2002

 19

15 males; mean age 56 yrs; PLP ≥ 6 mo; average pain intensity ≈ 6 on VAS; majority lower limb amputations; surgical amputations

Randomised, DB, cross‐over; 1 wk wash‐out; CGR

At 6 wks

Two in treatment group, non‐completion and protocol violation; three in placebo, non‐completion; consent withdrawal

 5

 9

Smith 2005

24

18 males; mean age 52 yrs; average pain intensity four NRS; patients with PLP and RLP; upper, lower limb amputations; vascular, traumatic, cancer, infections; at least 6 mo since amputation

Randomised, DB, cross‐over, 5 wk wash‐out; CGR

At 6 wks

Not described

4

7

Antidepressants

Amitriptyline

Robinson 2004

39

17 males; mean age 44 to 45 yrs; with PLP or RLP; upper, lower limb amputation; vascular, traumatic, cancer, infectious; pain at least 3 mos; pain intensity at 2 NRS

Randomised, DB, parallel

Central pharmacy randomisation

At 6 wks

Two due to side effects

5

9

Calcitonins

Jaeger 1992

21

12 males; median age 59 yrs; with severe PLP, 0 to 7 days after surgery; all except one are lower limb amputations; vascular, traumatic, malignant, infectious; pain intensity at least 3 NAS

Controlled clinical trial, cross‐over, 2 hr wash‐out; DB followed by open phase

Short‐term: 24 hr before and after treatment (double blind); long‐term: 6 mos, 1 to 2 yrs (open phase)

None in short‐term; total of eight patients in long‐term (open phase)

3

6

Eichenberger 2008

20

15 males, median age 57 yrs; mean pain intensity 4.32 on 10 cm VAS, average of 12.41 yrs PLP

Randomised, DB, cross‐over, time between infusions 48 hours; drawing of lots by person not involved in study

30, 60 min, 48 hrs after infusion

One from ketamine alone group did not get saline; two from placebo did not get heat, electrical stimulation

5

9

Opioids

Morphine

Huse 2001

12

10 males; mean age 50 yrs; PLP; upper, lower  limb amputations, pain intensity at least 3 VAS; average time since amputation 16 yrs

Randomised, cross‐over; 4‐wk DB phase, 1 to 2 wks wash‐out; long‐term phase (open) for responders; physician with no contact with patients randomised and kept code

Hourly for pain and side effect; weekly during 4 weeks of DB phase; long‐term 6, 12 mos (open)

None during double blind phase

4

8

Wu 2002

31

19 males; mean age 54 yrs; patients with persistent post amputation pains 6 mos; lower, upper limb amputations; time since amputation 81 mos

Randomised, DB, cross‐over, 24 hrs wash‐out; active‐placebo controlled; block randomisation

30 min after end of infusion

One due to absence of pain before start of infusions

5

9

Anaesthetics

Lidocaine

Wu 2002

31

19 males; mean age 54 yrs; patients with persistent post amputation pain > 6 mos; lower, upper limb amputations; time since amputation 81 mos

Randomised, DB, cross‐over, 24 hrs wash‐out; active‐placebo controlled; block randomisation

30 min after end of infusion

One due to absence of pain before start of infusions

5

9

Bupivacaine

Casale 2009

8

6 males; mean age 70 yrs;

PLP at least 6 mos; lower limb amputation; traumatic, vascular; average pain intensity ≈7

 Randomised, DB, cross‐over, 72 hrs wash‐out; CGR

60 min after injection

none

4

10

DB, double‐blind; CGR, computer‐generated randomisation; NAS, numerical analog scale; VAS, visual analog scale; NRS, numerical rating scale; PLP, phantom limb pain; RLP, residual limb pain; yr, year; yrs, years; wk, week; wks, weeks; hr, hour; hrs, hours; min, minutes; mo, month; mos, months; pt, point

Figuras y tablas -
Table 1. Summary of Characteristics of Included Studies
Table 2. Summary of Results

Author,

yr

Intervention

Treatment

Duration

FF‐up

Outcomes

Results

Over‐all

direction

of

efficacy

Adverse

events

NMDA

antagonist

 

 

 

 

 

 

 

Memantine

 

 

 

 

 

 

 

Maier 2003

 1) memantine

30 mg/d; oral

2) placebo

 3 weeks

At

end of

3 weeks

Pain intensity

11 pt NRS;

number of

participants

with ≥50% pain

reduction;NNT;

mood;disability;

adverse

events

No sig diff in

change in

pain level, in

number of

participants with

≥50% pain

relief; depres‐

sion scores;

disability indices

in 2 grps; over‐

all number

severe events

higher in

memantine

 ‐

vertigo,

tiredness,

headache,

nausea,

restlessness,

excitation,

cramps

Wieck 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

0‐100 VAS;

MEG

recording;

adverse

events

No sig diff in

change in pain

intensity,

cortical

reorganization

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 pt NRS;

ICI; ICF

No sig diff in

pain intensity;

enhanced ICI;

reduced ICF

 ‐

 Not

described

 Dextro‐

methorphan

 

 

 

 

 

 

 

Abraham 2003

 1)

dextromethor‐

phan

120 mg/d;oral

2)

dextromethor‐

phan

180 mg/d;oral

3) placebo

10 days

each

treatment

arm 

At

end of

10

days of

each

arm

Number of

patients with

≥50% pain

relief; feeling

of well‐being;

sedation

score;

adverse

events

Dextromethor‐

phan grps with

≥50% pain

relief; with signi‐

ficantly better

feeling of well‐

being scores;

with

significantly

lower sedation

scores

 +

none

reported

 Ketamine

 

 

 

 

 

 

 

Nikolajsen 1996

 1) ketamine

0.5 mg/kg

once, IV

infusion

2) placebo

45

minute

each

treatment

arm

At end

of IV

infusion

Pain intensity

0‐100 mm

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

patients with

≥50% pain

reduction on

10 cm VAS;

basal

sensory

assessment;

adverse

effects

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 mm 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 wks

of each

arm

Pain intensity

0‐10 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

0‐10 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);

7‐152

days,

weekly

(open

phase)

Pain intensity

0‐10 NAS in

open phase/

long‐term;

number of

patients 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 patients

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

patients with

≥50% pain

relief on 10

cm 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

treat‐

ment

phase

of 4

weeks

Pain intensity

10 cm VAS;

number of

patients 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

25‐50% pain

relief during

morphine; no

sig change in

perception and

pain thresholds;

significantly

lower attentional

performance

during

morphine;

scores on pain

experience

scale, depres‐

sion score,

WHYMPI, BSS

with no sig

relationship with

pain reduction;

2 of 3 with clear

cortical

reorganization

+

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

(diphenhydra‐

mine)

40

minutes

of IV

infusion

30

minutes

after

end of

infusion

Pain relief 0‐

100% numeric

scale; NNT for

30% pain

reduction;

satisfaction,

sedation

scores,

adverse

events

Sig dec in

phantom and

stump pain

intensity during

IV morphine;

NNT=2;

significantly

higher

satisfaction with

morphine;

no sig diff in

sedation

scores

+

sedation

(but no sig

diff with other

groups)

Anaesthetics

Lidocaine

Wu 2002

1) morphine

0.2 mg/kg, IV

infusion

2) lidocaine

4 mg/kg, IV

infusion

3) placebo

(diphenhydra‐

mine)

40

minutes

of IV

infusion

30

minutes

after

end of

infusion

Pain relief 0‐

100% numeric

scale; NNT for

30% pain

reduction;

satisfaction;

sedation

scores;

adverse

events

No sig dec in

PLP vs.

placebo; NNT=

3.8; 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

one

hour

 Pain intensity

0‐10 VAS;

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 patients

 +

 none

yr, year; d, day; mins, minutes; NRS, numerical rating scale; NNT, number needed to treat; VAS, visual analog scale; sig, significant; dec, decrease; inc, increase; grps, groups; grp, group; max, maximum; ICI, intracortical inhibition; ICF, intracortical facilitation; IV, intravenous; DB, double‐blind; MEG, magnetoencephalography; HADS, Hospital Anxiety and Depression Scale; BI, Barthel Index; NRS, numerical rating scale; CES‐D, Center Epidemiologic Depression Scale; FIM, Functional Independence Measure; CHART, Craig Handicap Assessment and Reporting Technique; NAS, numerical analog scale; WHYMPI, West‐Haven Yale Multidimensional Inventory; BSS, Brief Stress Scale; apain intensity; bmood, sleep, function; cpain intensity, mood, function, handicap, satisfaction

Figuras y tablas -
Table 2. Summary of Results
Comparison 1. memantine vs. placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

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 vs. placebo
Comparison 2. gabapentin vs. placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 change in pain intensity Show forest plot

2

Mean Difference (Fixed, 95% CI)

‐1.16 [‐1.94, ‐0.38]

Figuras y tablas -
Comparison 2. gabapentin vs. placebo