Scolaris Content Display Scolaris Content Display

Levetiracetam para el dolor neuropático en adultos

Collapse all Expand all

Resumen

Antecedentes

Los fármacos antiepilépticos se han usado en el manejo del dolor desde la década de los sesenta; algunos han demostrado eficacia en el tratamiento de diferentes afecciones de dolor neuropático. La eficacia del levetiracetam para el alivio del dolor neuropático no se ha examinado anteriormente.

Objetivos

Evaluar la eficacia analgésica y los eventos adversos del levetiracetam en adultos con trastornos de dolor neuropático crónico.

Métodos de búsqueda

Se hicieron búsquedas en el Registro Cochrane Central de Ensayos Controlados (Cochrane Central Register of Controlled Trials) (CENTRAL) (2014, número 6) (vía la Cochrane Library), MEDLINE, EMBASE, y en dos bases de datos de ensayos clínicos (ClinicalTrials.gov y la World Health Organisation Clinical Trials Registry Platform) hasta el 3 julio 2014, junto con listas de referencias de artículos y revisiones recuperados.

Criterios de selección

Se incluyeron estudios aleatorizados doble ciego de dos semanas de duración o más, que compararan levetiracetam con placebo u otro tratamiento activo en adultos con trastornos de dolor neuropático crónico. Los estudios debían tener un mínimo de diez participantes por brazo de tratamiento.

Obtención y análisis de los datos

De forma independiente, dos autores de la revisión extrajeron los datos sobre la eficacia y los eventos adversos y examinaron las cuestiones de calidad de los estudios. Se realizó el análisis con tres niveles de evidencia. El primer nivel de evidencia se obtuvo a partir de los datos que cumplían con los mejores estándares actuales y que poseían un riesgo de sesgo mínimo (resultado equivalente a una reducción significativa en la intensidad del dolor, análisis por intención de tratar sin imputación de los abandonos, al menos 200 participantes en la comparación, duración de ocho a 12 semanas, diseño paralelo), el segundo nivel de evidencia a partir de los datos que no cumplían con uno o más de estos criterios y que se consideró que poseían cierto riesgo de sesgo, pero con al menos 200 participantes en la comparación y el tercer nivel de evidencia, a partir de los datos que incluyeron menos de 200 participantes que se consideraron con alta probabilidad de sesgo o que utilizaran resultados de escasa utilidad clínica, o ambos.

Resultados principales

Se incluyeron seis estudios: cinco estudios pequeños, cruzados (cross‐over) con 174 participantes y un estudio de grupos paralelos con 170 participantes. Los participantes recibieron tratamiento con levetiracetam (2000 mg a 3000 mg diarios) o placebo durante entre cuatro y 14 semanas. Cada estudio incluyó participantes con un tipo diferente de dolor neuropático; dolor central debido a esclerosis múltiple, dolor después de una lesión de la médula espinal, polineuropatía dolorosa, dolor central después de un accidente cerebrovascular, neuralgia postherpética y dolor después de una mastectomía.

Ninguno de los estudios incluidos proporcionó evidencia de primer o segundo nivel. La calidad de la evidencia fue muy baja, y la misma disminuyó debido al tamaño pequeño de los brazos de tratamiento, y debido a que los estudios presentaron los resultados mediante el uso de la imputación de la última observación transferida (LOCF, por sus siglas en inglés) para los retiros o utilizando sólo a los participantes que finalizaron el estudio según el protocolo, cuando hubo más de un 10% de retiros. No hubo datos suficientes para realizar un análisis agrupado de eficacia sobre trastornos particulares de dolor neuropático, aunque los estudios individuales no revelaron efectos analgésicos del levetiracetam en comparación con placebo. Se agruparon los resultados de cualquier resultado considerado alivio sustancial del dolor (≥ 50% de reducción de la intensidad del dolor o respuestas"completas" o"buenas" en la escala de calificación verbal) para cuatro estudios con datos dicotómicos; las tasas de respuesta a través de diferentes tipos de dolor neuropático fueron similares con levetiracetam (10%) y placebo (12%), sin diferencias estadísticas (riesgos relativos 0,9; intervalo de confianza [IC] del 95%: 0,4 a 1,7).

Se combinaron los datos a través de diferentes trastornos para los eventos adversos y los retiros. Sobre la base de datos muy limitados, significativamente más participantes experimentaron un evento adverso con levetiracetam en comparación con placebo (número necesario a tratar para un evento perjudicial adicional [NND] 8,0 [IC del 95%: 4,6 a 32]). Hubo significativamente más retiros por eventos adversos con levetiracetam (NND 9,7 [6,7 a 18]).

Conclusiones de los autores

La cantidad de evidencia sobre el levetiracetam en los trastornos de dolor neuropático fue muy pequeña y la misma presentó sesgos potenciales debido a los métodos de análisis utilizados en los estudios. No hubo ninguna indicación de que el levetiracetam fuera efectivo para aliviar el dolor neuropático, aunque se asoció con un aumento de los participantes que experimentaron eventos adversos y que se retiraron debido a los eventos adversos.

PICOs

Population
Intervention
Comparison
Outcome

The PICO model is widely used and taught in evidence-based health care as a strategy for formulating questions and search strategies and for characterizing clinical studies or meta-analyses. PICO stands for four different potential components of a clinical question: Patient, Population or Problem; Intervention; Comparison; Outcome.

See more on using PICO in the Cochrane Handbook.

Resumen en términos sencillos

Levetiracetam para el dolor neuropático en adultos

El dolor neuropático puede surgir del daño a los nervios y la lesión al sistema nervioso central. Es diferente de los mensajes de dolor transmitidos a lo largo de los nervios sanos a partir del tejido dañado (una caída, o corte, o la artritis de la rodilla). El dolor neuropático se trata con fármacos diferentes que los administrados para el dolor que proviene del tejido dañado. Por lo general, los fármacos como el paracetamol o el ibuprofeno no son efectivos para el dolor neuropático, mientras que la medicación que a veces se utiliza para tratar la depresión o la epilepsia puede ser muy efectiva para algunos pacientes con dolor neuropático.

El levetiracetam es un fármaco normalmente usado para tratar la epilepsia. Algunos de estos fármacos también son útiles para tratar el dolor neuropático. Se realizaron búsquedas de los ensayos clínicos en los cuales el levetiracetam se usó para tratar el dolor neuropático. Se encontraron seis estudios en 344 participantes adultos con seis trastornos de dolor neuropático diferentes publicados hasta julio de 2014. Estos estudios fueron aleatorizados y doble ciego, lo cual generalmente significa que es posible confiar en los mismos. Sin embargo, todos presentaron uno o más problemas que podrían dar lugar a que los resultados parecieran mejores que los encontrados en la práctica. No hubo ningún beneficio del levetiracetam en ninguno de los seis trastornos. Más participantes experimentaron eventos adversos con levetiracetam (67 de 100) que con placebo (54 de 100) e interrumpieron el tratamiento con levetiracetam debido a los eventos adversos (13 de 100) en comparación con los que interrumpieron el placebo (dos de 100). Los eventos adversos fueron: cansancio, mareos, cefalea, estreñimiento y náuseas.

Hubo muy poca información, que fue de calidad inadecuada, para tener la seguridad de que el levetiracetam funciona como una medicación para el dolor en alguno de los trastornos de dolor neuropático investigados. Otros fármacos han mostrado ser efectivos en algunos de estos trastornos.

Authors' conclusions

Implications for practice

This review found no evidence to suggest that levetiracetam provides pain relief in any neuropathic pain condition. Studies were small, mostly of relatively short duration for a chronic condition, and were potentially subject to major bias. There are more effective medicines available for the more common neuropathic pain conditions (Kalso 2013;Lunn 2014; Moore 2013c; Wiffen 2013).

Implications for research

Reasonable levels of evidence exist for the benefit of other anti‐epileptic and antidepressant drugs in the treatment of chronic neuropathic pain (for example, 14 studies of pregabalin in 3680 participants (Moore 2009); nine studies of duloxetine in 2776 participants (Lunn 2014)). Although the evidence for levetiracetam was of very low quality, there does not appear to be any justification for continued research with the drug in neuropathic pain given the poor results for efficacy, and because other treatments with evidence of significant efficacy are available.

Summary of findings

Open in table viewer
Summary of findings for the main comparison.

Levetiracetam compared with placebo for neuropathic pain

Patient or population: neuropathic pain (6 studies in central neuropathic pain due to multiple sclerosis, spinal cord injury, polyneuropathy, central post‐stroke pain, postherpetic neuralgia, and post‐mastectomy pain)

Intervention: levetiracetam 2000 mg to 3000 mg daily

Comparison: placebo

Outcomes

Probable outcome with comparator (placebo)

Probable outcome with intervention

Relative effect
(95% CI)

No. of participants
and studies

Quality of the evidence
(GRADE)

Comments

At least 50% reduction in pain

9/57

9/59

Not calculated

2 studies, 59 participants

Very low

Small numbers of studies and participants, cross‐over studies, potential bias in analysis

At least 30% reduction in pain

12/57

11/59

Not calculated

2 studies, 59 participants

Very low

Small numbers of studies and participants, cross‐over studies, potential bias in analysis

Proportion below 30/100 mm on VAS

No data

Patient Global Impression of Change very much improved (patient global evaluation of pain relief complete or good)

6/86

4/86

Not calculated

3 studies, 86 participants

Very low

Small numbers of studies and participants, cross‐over studies, potential bias in analysis

Patient Global Impression of Change much or very much improved (patient global evaluation of pain relief complete, good or moderate)

8/86

13/86

Not calculated

3 studies, 86 participants

Very low

Small numbers of studies and participants, cross‐over studies, potential bias in analysis

Any measure of 'substantial' pain relief

14/119

12/121

Not calculated

4 studies, 121 participants

Very low

Small numbers of studies and participants, cross‐over studies, potential bias in analysis

Adverse event withdrawals

24 in 1000

130 in 1000

RR 4.9 (2.2 to 11)

NNH 9.7 (6.7 to 18)

6 studies, 334 participants in total

Very low

Small numbers of studies and participants

Serious adverse events

2/334

2/334

Not calculated

6 studies, 334 participants in total

Very low

Small numbers of studies and participants

Death

No deaths

Very low

Small numbers of studies and participants

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

CI: confidence interval; NNH: number needed to treat to harm; RR: risk ratio; VAS: visual analogue scale

Background

This review is based on a template for reviews of drugs used to relieve neuropathic pain. The aim is for all reviews to use the same methods, based on new criteria for what constitutes reliable evidence in chronic pain (Moore 2010a; Appendix 1). An overview review of antiepileptic drugs for treating neuropathic pain identified that no Cochrane review existed for levetiracetam (Wiffen 2013).

Description of the condition

The 2011 International Association for the Study of Pain definition of neuropathic pain is "pain caused by a lesion or disease of the somatosensory system" (Jensen 2011) based on a definition agreed at an earlier consensus meeting (Treede 2008). Neuropathic pain may be caused by nerve damage, but is often followed by changes in the central nervous system (CNS) (Moisset 2007). The genesis of neuropathic pain is complex (Apkarian 2011; Baron 2010; Baron 2012; Tracey 2011; von Hehn 2012), and neuropathic pain features can be found in patients with joint pain (Soni 2013). Many people with neuropathic pain conditions are disabled with significant levels of pain for many years. Chronic painful conditions comprise five of the 11 top‐ranking conditions for years lived with disability in 2010 (Vos 2012), and are responsible for considerable loss of quality of life, employment, and increased health costs (Moore 2013a).

In primary care in the UK the incidences, per 100,000‐person years observation, have been reported as 28 (95% confidence interval (CI) 27 to 30) for postherpetic neuralgia, 27 (95% CI 26 to 29) for trigeminal neuralgia, 0.8 (95% CI 0.6 to 1.1) for phantom limb pain and 21 (95% CI 20 to 22) for painful diabetic neuropathy (Hall 2008). These estimates are in accordance with a systematic review of epidemiological studies from a number of different countries (van Hecke 2014). In other studies the incidence of trigeminal neuralgia has been estimated at 4 in 100,000 per year (Katusic 1991; Rappaport 1994), while a study of facial pain in The Netherlands found incidences per 100,000‐person years of 12.6 for trigeminal neuralgia and 3.9 for postherpetic neuralgia (Koopman 2009). Estimates vary between studies, often because of small numbers of cases. A systematic review of chronic pain demonstrated that some neuropathic pain conditions, such as painful diabetic neuropathy, can be more common, with prevalence rates up to 400 per 100,000‐person years (McQuay 2007) illustrating how common the condition was as well as its chronicity. The prevalence of neuropathic pain was reported as being 3.3% in Austria (Gustorff 2008), 6.9% in France (Bouhassira 2008), as high as 8% in the UK (Torrance 2006), about 7% in a systematic review of studies published since 2000 (Moore 2013a), and 7% to 10% in a systematic review of epidemiological studies published between 1966 and 2012 (van Hecke 2014). The prevalence of some types of neuropathic pain, such as diabetic neuropathy and post surgical chronic pain (which is often neuropathic in origin), are increasing (Hall 2008).

Neuropathic pain is difficult to treat effectively, with only a minority of individuals experiencing a clinically relevant benefit from any one intervention. A multidisciplinary approach is now advocated, with pharmacological interventions being combined with physical or cognitive interventions, or both. Conventional analgesics are usually not effective. Some patients may derive some benefit from a topical lidocaine patch or low concentration topical capsaicin, though evidence about benefits is uncertain (Derry 2012; Khaliq 2007). High concentration topical capsaicin may benefit some patients with postherpetic neuralgia (Derry 2013). Treatment is often by so‐called unconventional analgesics, such as antidepressants like duloxetine and amitriptyline (Lunn 2014; Moore 2012a; Moore 2013d) or antiepileptics like gabapentin or pregabalin (Moore 2009; Moore 2011). While treatment guidelines have general similarities based on the evidence available, they are not always consistent with one another (O'Connor 2009). The proportion of patients who achieve worthwhile,meaningful pain relief (typically at least 50% pain intensity reduction (Moore 2013b)) is small, generally 10% to 25% more than with placebo, with numbers needed to treat to benefit (NNT) usually between 4 and 10 (Moore 2013c).

Description of the intervention

Levetiracetam is an antiepileptic drug that is related to piracetam – a drug used to treat cortical reflex myoclonus (a type of epilepsy that originates in the cerebral cortex). It is available as oral tablets of 250 mg, 500 mg and 1 g. In addition both an oral solution and an intravenous (IV) infusion are available.

How the intervention might work

The mode of action of levetiracetam has not been fully resolved but is thought to work via gamma‐aminobutyric acid (GABA) A receptors. Its mechanism of action is considered to be different from all other antiepileptic drugs (SPC 2013; Wakita 2013). A detailed description of the mode of action and evidence concerning the use of levetiracetam in epilepsy is available in another Cochrane review (Mbzivo 2012). Studies exist that have investigated the potential role of levetiracetam in neuropathic pain treatment.

Why it is important to do this review

Levetiracetam is not commonly prescribed for neuropathic pain and it is not licensed for the treatment of neuropathic pain in the UK or USA, but the potential for benefit from this drug needs to be investigated. This review is one of a series of reviews covering the role of antiepileptics in neuropathic pain, and will be included in an overview review (Wiffen 2013).

The standards used to assess evidence in chronic pain trials have changed substantially, with particular attention being paid to trial duration, withdrawals, and statistical imputation following withdrawal, all of which can substantially alter estimates of efficacy. The most important change is the move from using average pain scores, or average change in pain scores, to the number of patients who have a large decrease in pain (by at least 50%); this level of pain relief has been shown to correlate with improvements in comorbid symptoms, function, and quality of life. These standards are set out in the Cochrane Pain, Palliative and Supportive Care Group's Author and Referee Guidance for pain studies (AUREF 2012).

This Cochrane review will assess evidence in ways that make both statistical and clinical sense, and will use developing criteria for what constitutes reliable evidence in chronic pain (Moore 2010a; Moore 2012b). Studies included and analysed will need to meet a minimum of reporting quality (blinding, randomisation), validity (duration, dose and timing, diagnosis, outcomes, etc) and size (ideally at least 400 participants in a comparison in which the NNT is four or above (Moore 1998)). This does set high standards and marks a departure from how reviews have been done previously.

Objectives

  1. To assess the analgesic efficacy of levetiracetam for chronic neuropathic pain in adults.

  2. To assess the adverse events associated with the clinical use of levetiracetam for chronic neuropathic pain.

Methods

Criteria for considering studies for this review

Types of studies

We included studies if they were randomised controlled trials (RCTs) with double‐blind assessment of participant outcomes following two weeks of treatment or longer, although the emphasis of the review was on studies of eight weeks or longer. We required full journal publication, with the exception of online clinical trial results summaries of otherwise unpublished clinical trials and abstracts with sufficient data for analysis. We did not include short abstracts (usually meeting reports). We excluded studies that were non‐randomised, studies of experimental pain, case reports and clinical observations.

Types of participants

Studies included adults aged 18 years and above. Participants could have one or more of a wide range of chronic neuropathic pain conditions but we specifically searched for and included:

  • painful diabetic neuropathy;

  • postherpetic neuralgia;

  • trigeminal neuralgia;

  • phantom limb pain;

  • postoperative or traumatic neuropathic pain;

  • complex regional pain syndrome (CRPS), Type I and Type II;

  • cancer‐related neuropathy;

  • human immunodeficiency virus (HIV) neuropathy;

  • spinal cord injury.

If studies had included participants with more than one type of neuropathic pain we would have analysed results according to the primary condition.

Types of interventions

Levetiracetam at any dose, by any route, administered for the relief of neuropathic pain and compared to placebo or any active comparator.

Types of outcome measures

We anticipated that studies would use a variety of outcome measures, with the majority of studies using standard subjective scales (numerical rating scale (NRS) or visual analogue scale (VAS)) for pain intensity or pain relief, or both. We were particularly interested in Initiative on Methods, Measurement, and Pain Assessment in Clinical Trials (IMMPACT) definitions for moderate and substantial benefit in chronic pain studies (Dworkin 2008). These are defined as at least 30% pain relief over baseline (moderate), at least 50% pain relief over baseline (substantial), much or very much improved on Patient Global Impression of Change (PGIC) (moderate), and very much improved on PGIC (substantial). These outcomes are different from those used in most earlier reviews, concentrating as they do on dichotomous outcomes where pain responses do not follow a normal (Gaussian) distribution. People with chronic pain desire high levels of pain relief, ideally more than 50%, and with pain not worse than mild (O'Brien 2010).

We have included a 'Summary of findings' table as set out in the author guide (AUREF 2012). The 'Summary of findings' table includes outcomes of at least 50% and at least 30% pain intensity reduction, PGIC, adverse event withdrawals, serious adverse events and death.

Primary outcomes

  1. Participant‐reported pain relief of 30% or greater

  2. Participant‐reported pain relief of 50% or greater

  3. PGIC much or very much improved

  4. PGIC very much improved

Secondary outcomes

  1. Any pain‐related outcome indicating some improvement

  2. Participants experiencing any adverse event

  3. Participants experiencing any serious adverse event. Serious adverse events typically include any untoward medical occurrence or effect that at any dose results in death, is life‐threatening, requires hospitalisation or prolongation of existing hospitalisation, results in persistent or significant disability or incapacity, is a congenital anomaly or birth defect, is an ‘important medical event’ that may jeopardise the patient, or may require an intervention to prevent one of the above characteristics/consequences

  4. Specific adverse events, particularly somnolence and dizziness

  5. Withdrawals due to adverse events

  6. Withdrawals due to lack of efficacy

Search methods for identification of studies

Electronic searches

We searched the following databases without language restrictions:

  • the Cochrane Central Register of Controlled Trials (CENTRAL) (2014, Issue 6) (via the Cochrane Library);

  • MEDLINE (via Ovid) from 1946 to 3 July 2014;

  • EMBASE (via Ovid) from 1974 to 3 July 2014.

The search strategies for MEDLINE, EMBASE and CENTRAL are in Appendix 2, Appendix 3, and Appendix 4.

Searching other resources

We reviewed the bibliographies of any RCTs identified and review articles, contacted known experts in the field, and searched clinical trial databases (ClinicalTrials.gov (ClinicalTrials.gov) and the World Health Organization (WHO) International Clinical Trials Registry Platform (ICTRP) (http://apps.who.int/trialsearch/)) to identify additional published or unpublished data. We did not contact investigators or study sponsors.

Data collection and analysis

The intention was to perform separate analyses according to particular neuropathic pain conditions. Analyses combining different neuropathic pain conditions would be done for exploratory purposes only.

Selection of studies

Two authors (PW and SD) independently determined eligibility by reading the abstract of each study identified by the search. Independent authors eliminated studies that clearly did not satisfy inclusion criteria, and we obtained full copies of the remaining studies. Two review authors (PW and SD) read these studies independently and reached agreement by discussion. In the event of disagreement, a third author would adjudicate. We did not anonymise the studies in any way before assessment. A Preferred Reporting Items for Systematic Reviews and Meta‐Analyses (PRISMA) flow chart shows the status of identified studies (Figure 1).


Study flow diagram.

Study flow diagram.

Data extraction and management

Two review authors independently extracted data using a standard form and checked for agreement before entry into RevMan (RevMan 2012). We included information about the pain condition and number of participants treated, drug and dosing regimen, study design (placebo or active control), study duration and follow‐up, analgesic outcome measures and results, withdrawals and adverse events (participants experiencing any adverse event, or serious adverse event).

Assessment of risk of bias in included studies

We used the Oxford Quality Score as the basis for inclusion (Jadad 1996), limiting inclusion to studies that were randomised and double‐blind as a minimum.

Two authors (PW and SD) independently assessed the risk of bias for each study, using the criteria outlined in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011) and adapted from those used by the Cochrane Pregnancy and Childbirth Group, with any disagreements resolved by discussion. We assessed the following for each study:

  1. Random sequence generation (checking for possible selection bias). We assessed the method used to generate the allocation sequence as: low risk of bias (any truly random process, for example random number table; computer random number generator); unclear risk of bias (method used to generate sequence not clearly stated). We excluded studies using a non‐random process (for example, odd or even date of birth; hospital or clinic record number).

  2. Allocation concealment (checking for possible selection bias). The method used to conceal allocation to interventions prior to assignment determines whether intervention allocation could have been foreseen in advance of, or during recruitment, or changed after assignment. We assessed the methods as: low risk of bias (for example, telephone or central randomisation; consecutively numbered sealed opaque envelopes); unclear risk of bias (method not clearly stated). We excluded studies that did not conceal allocation (for example, open list).

  3. Blinding of outcome assessment (checking for possible detection bias). We assessed the methods used to blind study participants and outcome assessors from knowledge of which intervention a participant received. We assessed the methods as: low risk of bias (study stated that it was blinded and describes the method used to achieve blinding, for example, identical tablets; matched in appearance and smell); unclear risk of bias (study stated that it was blinded but does not provide an adequate description of how it was achieved). We excluded studies that were not double‐blind.

  4. Incomplete outcome data (checking for possible attrition bias due to the amount, nature and handling of incomplete outcome data). We assessed the methods used to deal with incomplete data as: low risk (< 10% of participants did not complete the study and/or used ‘baseline observation carried forward’ analysis); unclear risk of bias (used 'last observation carried forward' analysis); high risk of bias (used 'completer' analysis).

  5. Size of study (checking for possible biases confounded by small size). We assessed studies as being at low risk of bias (≥ 200 participants per treatment arm); unclear risk of bias (50 to 199 participants per treatment arm); high risk of bias (< 50 participants per treatment arm).

Measures of treatment effect

We planned to calculate NNTs as the reciprocal of the absolute risk reduction (ARR) (McQuay 1998). For unwanted effects, the NNT becomes the number needed to treat to harm (NNH) and is calculated in the same manner. We planned to use dichotomous data to calculate risk ratio (RR) with 95% confidence intervals (CI) using a fixed‐effect model unless significant statistical heterogeneity was found (see below). Continuous data were not used in analyses.

Dealing with missing data

We used intention‐to‐treat (ITT) analysis where the ITT population consists of participants who were randomised, took at least one dose of the assigned study medication, and provided at least one post‐baseline assessment. Missing participants were assigned zero improvement wherever possible.

Assessment of heterogeneity

We planned to deal with clinical heterogeneity by combining studies that examined similar conditions, and to assess statistical heterogeneity visually (L'Abbé 1987) and with the use of the I² statistic. If I² was greater than 50%, we would consider possible reasons.

Assessment of reporting biases

The aim of this review was to use dichotomous data of known utility (Moore 2010d). The review does not depend on what authors of the original studies chose to report or not. We would extract and use continuous data, which probably poorly reflect efficacy and utility, if useful for illustrative purposes only.

We planned to assess publication bias using a method designed to detect the amount of unpublished data with a null effect required to make any result clinically irrelevant (usually taken to mean NNT values of 10 or higher) (Moore 2008).

Data synthesis

We planned to use a fixed‐effect model for meta‐analysis. A random‐effects model for meta‐analysis would have been used if there was significant clinical heterogeneity and it was considered appropriate to combine studies.

We planned to analyse efficacy data for each painful condition in three tiers, according to outcome and freedom from known sources of bias.

  • The first tier used data meeting current best standards, where studies reported the outcome of at least 50% pain intensity reduction over baseline (or its equivalent), without the use of last observation carried forward (LOCF) or other imputation method for dropouts, reported an intention‐to‐treat (ITT) analysis, lasted eight or more weeks, had a parallel‐group design, and had at least 200 participants (preferably at least 400) in the comparison (Moore 1998; Moore 2010a; Moore 2012a). These top‐tier results would be reported first.

  • The second tier used data from at least 200 participants but where one or more of the above conditions were not met (for example reporting at least 30% pain intensity reduction, using LOCF or a completer analysis, or lasting four to eight weeks).

  • The third tier of evidence relates to data from studies including fewer than 200 participants, or where there were expected to be significant problems because, for example, of very short duration studies of less than four weeks, where there was major heterogeneity between studies, or where there were shortcomings in allocation concealment, attrition, or incomplete outcome data. For this third tier of evidence, no data synthesis is reasonable, and may be misleading, but an indication of beneficial effects might be possible.

We pooled adverse event data from different neuropathic pain conditions because there was no reason to believe that adverse responses would differ, and to provide a larger data set for analysis.

Subgroup analysis and investigation of heterogeneity

We planned efficacy analyses to be according to individual painful conditions, because placebo response rates with the same outcome can vary between conditions, as can the drug‐specific effects (Moore 2009). Analysis that pooled data from different conditions was carried out where there were insufficient data to analyse conditions separately, and only to give an indication of direction of effect.

Sensitivity analysis

We planned no sensitivity analysis because the evidence base was known to be too small to allow reliable analysis. We would examine details of dose escalation schedules, if available, in the unlikely situation that this could provide some basis for a sensitivity analysis.

Results

Description of studies

Results of the search

We identified 66 potentially relevant records in MEDLINE, 341 in EMBASE, and 27 in CENTRAL. Searches of clinical trials databases identified a further seven potential studies. After reading the abstracts, we read six articles and two clinical trial summaries in full. One of the two clinical trial summaries had insufficient data to determine whether it was randomised study and it was placed in Characteristics of studies awaiting classification (NCT00156689); the principal investigator is unable to provide any data (personal communication). The other clinical trial summary provided little detail (NCT00160511) (Figure 1).

Included studies

We included six studies (344 participants), each enrolling participants with a different type of neuropathic pain: central pain due to multiple sclerosis (Falah 2012), pain following spinal cord injury (Finnerup 2009), painful polyneuropathy (Holbech 2011), central post‐stroke pain (Jungehulsing 2013), postherpetic neuralgia (NCT00160511), and post‐mastectomy pain (Vilholm 2008).

All participants had experienced their pain for at least three months, and the intensity at study entry was moderate or severe (generally 4/10 to 9/10). The mean age in included studies ranged from 47 to 70 years, and all included both men and women except one in post‐mastectomy pain, which included only women (Vilholm 2008). Exclusion criteria for all studies (except NCT00160511, which did not provide information) included causes of pain other than that specified, previous allergic reaction or severe adverse events to levetiracetam or a similar medication, pregnancy or lactation, and severe terminal illness or other condition that would interfere with the study.

All the studies were placebo‐controlled. Five used a cross‐over design, and one a parallel group design (NCT00160511). Treatment periods were four to eight weeks, with a one‐ or two‐week washout between periods for cross‐over studies, and 14 weeks (four‐week up titration, eight‐week maintenance, two‐week taper) for the parallel group study. Previous medication for neuropathic pain was tapered and stopped completely during a pre‐study period of one to four weeks in Falah 2012, Holbech 2011, and Vilholm 2008, but some medication was continued (if it was stable and remained unchanged) in Finnerup 2009 and Jungehulsing 2013. NCT00160511 included a baseline period, but there was no information on washout of existing medication or permitted medication. Levetiracetam was titrated over approximately two weeks in cross‐over studies and four weeks in the parallel group study, from 500 mg or 1000 mg daily to a maximum of 3000 mg daily, and taken twice a day. All except two studies (NCT00160511; Vilholm 2008) stated that the dose could be reduced to a minimum of 2000 mg daily or to the previous tolerated dose in the event of intolerable side effects. Rescue medication was specified as 3000 mg or 4000 mg paracetamol daily, sometimes with the addition of tramadol 50 mg, in all but two studies (Jungehulsing 2013; NCT00160511).

Further details are provided in the Characteristics of included studies table. Study drugs were provided in all of the included studies by UCB Pharma, who also provided some financial support.

Excluded studies

We excluded one study because it was single blind (Rossi 2009).

Risk of bias in included studies

Comments on potential biases in individual studies are reported in the Risk of bias section of the Characteristics of included studies table. The findings are displayed in Figure 2 and Figure 3; no sensitivity analysis was undertaken. The greatest risk of bias came from small study size.


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

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.

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

Allocation

All except one of the studies (NCT00160511) adequately described the method used to generate the random number sequence, and all but two also described how the allocation was concealed (Jungehulsing 2013; NCT00160511).

Blinding

All the studies were double blind, and all but two adequately described how the blinding was achieved (Jungehulsing 2013; NCT00160511).

Incomplete outcome data

Four cross‐over studies stated that they used last observation carried forward (LOCF) imputation for participants who withdrew from the study, and we judged them at unknown risk of bias (Falah 2012; Finnerup 2009; Holbech 2011; Vilholm 2008). One cross‐over study reported a per protocol analysis and had more than 10% withdrawals, so we judged it at high risk of bias (Jungehulsing 2013). For some outcomes, analyses included only those participants who provided data for both phases of the cross‐over. None of the cross‐over studies reported efficacy results separately for the first treatment phase.

The parallel group study had high withdrawal rates (33% with levetiracetam and 22% with placebo) and did not report how these withdrawals were handled in the analysis of mean data (NCT00160511). All participants were included in analyses of adverse events and withdrawals.

Other potential sources of bias

Five of the studies were small, with fewer than 50 participants per treatment arm, and we judged them at high risk of bias. The remaining study, with 84 and 86 participants in the treatment arms, was judged at unclear risk of bias.

Effects of interventions

See: Summary of findings for the main comparison

None of the included studies provided first or second tier evidence. We downgraded the evidence was because of the small size of the treatment arms and because studies reported results using LOCF imputation for withdrawals or reported a per protocol analysis where there were greater than 10% withdrawals.

NCT00160511 did not report any dichotomous data and did not contribute to any efficacy analysis.

Results in individual studies are in Appendix 5 (efficacy) and Appendix 6 (adverse events and withdrawals).

Third tier evidence

Participant‐reported pain relief of 30% or greater, and 50% or greater

Two studies reported these outcomes.

In Finnerup 2009, 3/34 participants treated with levetiracetam, and 4/32 with placebo experienced at least 33% reduction in pain at the end of the five‐week treatment periods; 1/34 with levetiracetam and 1/32 with placebo experienced at least 50% pain reduction over five weeks.

In Vilholm 2008, 8/25 participants experienced at least 50% pain reduction over the four‐week treatment period with both levetiracetam and placebo.

Patient Global Impression of Change (PGIC) 'much or very much improved' and 'very much improved'

No studies reported these outcomes using the standard 5‐point scale. However, three studies used a 6‐point verbal rating scale to measure participants' pain relief at the end of the treatment periods (Falah 2012; Finnerup 2009; Holbech 2011): ‘complete’(6), ‘good’ (5), ‘moderate’ (4), ‘slight’ (3), ‘none’ (2) or ‘worse’ (1). While this was not a predefined outcome we considered it equivalent to PGIC very much improved when the result reported was 'complete' or 'good', and PGIC much or very much improved when the result reported was 'moderate', 'complete' or 'good' (Moore 2013b).

In Falah 2012, no participants experienced complete relief, 1/27 reported 'good' relief with both levetiracetam and placebo, and 4/27 reported 'moderate' relief with levetiracetam compared to 1/27 with placebo.

In Finnerup 2009, no participants experienced complete relief, 1/24 reported 'good' relief with both levetiracetam and placebo, and 2/24 reported 'moderate' relief with levetiracetam compared to 0/24 with placebo.

In Holbech 2011, 1/35 participants experienced complete relief with levetiracetam and none with placebo, 1/35 reported 'good' relief with levetiracetam and 4/35 with placebo, and 3/35 reported 'moderate' relief with levetiracetam compared to 1/35 with placebo.

Analysis 1.1 shows the results for outcomes measuring substantial pain relief (≥ 50% pain intensity reduction or ‘complete’ or ‘good’ responses on the verbal rating scale) for the four studies with dichotomous data. Where studies reported both outcomes, we have used ≥ 50% pain intensity reduction preferentially. The overall response rate across these different types of neuropathic pain was similar with levetiracetam (10%) and placebo (12%), with no individual study significantly different from placebo (Figure 4). There was no statistical difference between levetiracetam and placebo (RR 0.9; 95% CI 0.4 to1.7).


Forest plot of comparison: 1 Levetiriacetam versus placebo, outcome: 1.1 Participants with substantial pain relief (≥50% pain intensity reduction, or complete or good pain relief).

Forest plot of comparison: 1 Levetiriacetam versus placebo, outcome: 1.1 Participants with substantial pain relief (≥50% pain intensity reduction, or complete or good pain relief).

Any pain‐related outcome indicating some improvement

All the studies reported that the mean or median pain scores at the end of each treatment period did not differ between levetiracetam and placebo.

There was no evidence that levetiracetam provided better pain relief than placebo.

Use of rescue medication

Use of rescue medication was not a prespecified outcome in the protocol, but was reported in four studies. These data are analysed for completeness.

In the four studies that specified use of rescue medication, there was no difference in the mean number of tablets of paracetamol and tramadol (where allowed) consumed per week (Falah 2012; Finnerup 2009; Holbech 2011; Vilholm 2008), and usage was unchanged from the baseline period. During the baseline period and in both treatment arms, there was a very wide range in the number of tablets used.

Adverse events

We combined data from different neuropathic pain conditions for adverse events.

Participants experiencing any adverse event

Five studies reported the number of participants who experienced at least one adverse event during the treatment period (Falah 2012; Finnerup 2009; Holbech 2011; NCT00160511; Vilholm 2008). Combining the studies across conditions, 136/204 (67%) of participants experienced an adverse event with levetiracetam, and 111/205 (54%) with placebo. The risk ratio (RR) was 1.2 (95% confidence interval (CI) 1.1 to 1.5), and the number needed to treat for an additional harmful event (NNH) was 8.0 (4.6 to 32) (Analysis 1.2).

Where reported, the intensity of adverse events was mostly moderate or severe with levetiracetam, and mild or moderate with placebo. With levetiracetam the most common events were fatigue or tiredness, dizziness, headache, constipation, and nausea. With placebo the most common events were tiredness, headache, nausea, and constipation or abdominal discomfort.

Serious adverse events

Two serious adverse events were reported in both treatment arms of the parallel group study; none were judged related to study medication by the study investigators.

Deaths

There were no deaths reported in the studies.

Withdrawals

All studies reported on withdrawals for both treatment arms in participants who took at least one dose of study medication.

All cause withdrawals

Combining studies across conditions, a total of 59/251 (23%) of participants withdrew from treatment with levetiracetam, and 31/249 (12%) from treatment with placebo. The RR was 1.9 (95% CI 1.3 to 2.8), and the NNH was 9.1 (5.7 to 23) (Analysis 1.3).

Adverse events withdrawals

Combining studies across conditions, 32/251 (13%) of participants withdrew from treatment with levetiracetam, and 6/249 (2%) from treatment with placebo because of adverse events. The RR was 4.9 (95% CI 2.2 to 11), and the NNH was 9.7 (6.7 to 18) (Analysis 1.4; Figure 5).


Forest plot of comparison: 1 Levetiriacetam versus placebo, outcome: 1.4 Adverse event withdrawal.

Forest plot of comparison: 1 Levetiriacetam versus placebo, outcome: 1.4 Adverse event withdrawal.

Lack of efficacy withdrawals

Combining studies across conditions, 16/251 (6%) of participants withdrew from treatment with levetiracetam, and 12/249 (5%) from treatment with placebo because of lack of efficacy. The RR was 1.3 (95% CI 0.66 to 2.5). The NNH was not calculated (Analysis 1.5).

Discussion

Summary of main results

We found six studies enrolling 344 participants with chronic neuropathic pain of different origins: central pain due to multiple sclerosis, spinal cord injury, polyneuropathy, central post‐stroke pain, postherpetic neuralgia, and post‐mastectomy pain. All studies compared levetiracetam with placebo; five used a cross‐over design, and one a parallel group design.

No first or second tier evidence was available. No pooling of efficacy data was possible, but third tier evidence in individual studies did not indicate any improvement in pain relief with levetiracetam compared with placebo. Combining studies across pain conditions, participants taking levetiracetam were more likely to experience any adverse event during treatment (NNH 8.0), and the intensity of adverse events was reported as moderate or severe with levetiracetam, and mild to moderate with placebo. There were significantly more adverse event withdrawals with levetiracetam. Two serious adverse events were reported with both levetiracetam and placebo, all of which were judged unrelated to study medication by the investigators. There were no deaths in the studies. See summary of findings Table for the main comparison.

Overall completeness and applicability of evidence

Levetiracetam was tested in small numbers of people with six different types of neuropathic pain that did not include some of the most common types, such as painful diabetic neuropathy. The results here cannot reliably be extrapolated to other neuropathic pain conditions.

Treatment duration in most of the included studies was four to six weeks, and although short‐term studies (less than six weeks) may not accurately predict longer term efficacy in chronic conditions, it is very likely that some analgesic effect would be seen by four weeks if the intervention was effective in the conditions investigated. The single study of longer duration in postherpetic neuralgia also failed to show any benefit of levetiracetam based on group mean data.

The included studies were underpowered to provide reliable information relating to tolerability and safety.

Quality of the evidence

Reporting quality in the studies was generally poor by current standards. While all the studies were randomised and double‐blind, none provided data that met predefined criteria for first or second tier analysis. The studies were small, with only one having more than 50 participants in any treatment arm. Five used a cross‐over design without separate reporting of first period data, and reported only on participants who provided data for both phases of treatment. The single, larger parallel group study had a high rate of withdrawals and did not report how these were handled in the efficacy analysis of group mean data.

Potential biases in the review process

We carried out a broad search for studies, and think it is unlikely that significant amounts of data remain unknown to us.

Five of the included studies used a cross‐over design. The degree of exaggeration of treatment effects in cross‐over trials compared to parallel‐group designs, as has been seen in some circumstances (Khan 1996), is unclear but in itself is unlikely to be the source of major bias (Elbourne 2002). However, these studies reported results for both treatment periods combined, and only for participants who completed at least part of each treatment period, with last observation carried forward imputation for withdrawals, which is likely to overestimate efficacy.

Agreements and disagreements with other studies or reviews

We are not aware of any other reviews of levetiracetam for relief of neuropathic pain. Levetiracetam has been identified as a drug that should not been used for treating neuropathic pain because of a lack of evidence (NICE 2013).

Study flow diagram.
Figures and Tables -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figures and Tables -
Figure 2

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

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figures and Tables -
Figure 3

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

Forest plot of comparison: 1 Levetiriacetam versus placebo, outcome: 1.1 Participants with substantial pain relief (≥50% pain intensity reduction, or complete or good pain relief).
Figures and Tables -
Figure 4

Forest plot of comparison: 1 Levetiriacetam versus placebo, outcome: 1.1 Participants with substantial pain relief (≥50% pain intensity reduction, or complete or good pain relief).

Forest plot of comparison: 1 Levetiriacetam versus placebo, outcome: 1.4 Adverse event withdrawal.
Figures and Tables -
Figure 5

Forest plot of comparison: 1 Levetiriacetam versus placebo, outcome: 1.4 Adverse event withdrawal.

Comparison 1 Levetiriacetam versus placebo, Outcome 1 Participants with substantial pain relief (≥50% pain intensity reduction, or complete or good pain relief).
Figures and Tables -
Analysis 1.1

Comparison 1 Levetiriacetam versus placebo, Outcome 1 Participants with substantial pain relief (≥50% pain intensity reduction, or complete or good pain relief).

Comparison 1 Levetiriacetam versus placebo, Outcome 2 Participants with at least one adverse event.
Figures and Tables -
Analysis 1.2

Comparison 1 Levetiriacetam versus placebo, Outcome 2 Participants with at least one adverse event.

Comparison 1 Levetiriacetam versus placebo, Outcome 3 All cause withdrawal.
Figures and Tables -
Analysis 1.3

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

Comparison 1 Levetiriacetam versus placebo, Outcome 4 Adverse event withdrawal.
Figures and Tables -
Analysis 1.4

Comparison 1 Levetiriacetam versus placebo, Outcome 4 Adverse event withdrawal.

Comparison 1 Levetiriacetam versus placebo, Outcome 5 Lack of efficacy withdrawal.
Figures and Tables -
Analysis 1.5

Comparison 1 Levetiriacetam versus placebo, Outcome 5 Lack of efficacy withdrawal.

Levetiracetam compared with placebo for neuropathic pain

Patient or population: neuropathic pain (6 studies in central neuropathic pain due to multiple sclerosis, spinal cord injury, polyneuropathy, central post‐stroke pain, postherpetic neuralgia, and post‐mastectomy pain)

Intervention: levetiracetam 2000 mg to 3000 mg daily

Comparison: placebo

Outcomes

Probable outcome with comparator (placebo)

Probable outcome with intervention

Relative effect
(95% CI)

No. of participants
and studies

Quality of the evidence
(GRADE)

Comments

At least 50% reduction in pain

9/57

9/59

Not calculated

2 studies, 59 participants

Very low

Small numbers of studies and participants, cross‐over studies, potential bias in analysis

At least 30% reduction in pain

12/57

11/59

Not calculated

2 studies, 59 participants

Very low

Small numbers of studies and participants, cross‐over studies, potential bias in analysis

Proportion below 30/100 mm on VAS

No data

Patient Global Impression of Change very much improved (patient global evaluation of pain relief complete or good)

6/86

4/86

Not calculated

3 studies, 86 participants

Very low

Small numbers of studies and participants, cross‐over studies, potential bias in analysis

Patient Global Impression of Change much or very much improved (patient global evaluation of pain relief complete, good or moderate)

8/86

13/86

Not calculated

3 studies, 86 participants

Very low

Small numbers of studies and participants, cross‐over studies, potential bias in analysis

Any measure of 'substantial' pain relief

14/119

12/121

Not calculated

4 studies, 121 participants

Very low

Small numbers of studies and participants, cross‐over studies, potential bias in analysis

Adverse event withdrawals

24 in 1000

130 in 1000

RR 4.9 (2.2 to 11)

NNH 9.7 (6.7 to 18)

6 studies, 334 participants in total

Very low

Small numbers of studies and participants

Serious adverse events

2/334

2/334

Not calculated

6 studies, 334 participants in total

Very low

Small numbers of studies and participants

Death

No deaths

Very low

Small numbers of studies and participants

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

CI: confidence interval; NNH: number needed to treat to harm; RR: risk ratio; VAS: visual analogue scale

Figures and Tables -
Comparison 1. Levetiriacetam versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants with substantial pain relief (≥50% pain intensity reduction, or complete or good pain relief) Show forest plot

4

240

Risk Ratio (M‐H, Fixed, 95% CI)

0.85 [0.43, 1.69]

2 Participants with at least one adverse event Show forest plot

5

409

Risk Ratio (M‐H, Fixed, 95% CI)

1.24 [1.06, 1.45]

3 All cause withdrawal Show forest plot

6

501

Risk Ratio (M‐H, Fixed, 95% CI)

1.90 [1.28, 2.81]

4 Adverse event withdrawal Show forest plot

6

500

Risk Ratio (M‐H, Fixed, 95% CI)

4.90 [2.17, 11.09]

5 Lack of efficacy withdrawal Show forest plot

6

500

Risk Ratio (M‐H, Fixed, 95% CI)

1.29 [0.66, 2.52]

Figures and Tables -
Comparison 1. Levetiriacetam versus placebo