Scolaris Content Display Scolaris Content Display

Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia

Contraer todo Desplegar todo

Abstract

disponible en

Background

Duloxetine is a balanced serotonin and noradrenaline reuptake inhibitor licensed for the treatment of major depressive disorders, urinary stress incontinence and the management of neuropathic pain associated with diabetic peripheral neuropathy. A number of trials have been conducted to investigate the use of duloxetine in neuropathic and nociceptive painful conditions. This is the first update of a review first published in 2010.

Objectives

To assess the benefits and harms of duloxetine for treating painful neuropathy and different types of chronic pain.

Search methods

On 19th November 2013, we searched The Cochrane Neuromuscular Group Specialized Register, CENTRAL, DARE, HTA, NHSEED, MEDLINE, and EMBASE. We searched ClinicalTrials.gov for ongoing trials in April 2013. We also searched the reference lists of identified publications for trials of duloxetine for the treatment of painful peripheral neuropathy or chronic pain.

Selection criteria

We selected all randomised or quasi‐randomised trials of any formulation of duloxetine, used for the treatment of painful peripheral neuropathy or chronic pain in adults.

Data collection and analysis

We used standard methodological procedures expected by The Cochrane Collaboration.

Main results

We identified 18 trials, which included 6407 participants. We found 12 of these studies in the literature search for this update. Eight studies included a total of 2728 participants with painful diabetic neuropathy and six studies involved 2249 participants with fibromyalgia. Three studies included participants with depression and painful physical symptoms and one included participants with central neuropathic pain. Studies were mostly at low risk of bias, although significant drop outs, imputation methods and almost every study being performed or sponsored by the drug manufacturer add to the risk of bias in some domains. Duloxetine at 60 mg daily is effective in treating painful diabetic peripheral neuropathy in the short term, with a risk ratio (RR) for ≥ 50% pain reduction at 12 weeks of 1.73 (95% CI 1.44 to 2.08). The related NNTB is 5 (95% CI 4 to 7). Duloxetine at 60 mg daily is also effective for fibromyalgia over 12 weeks (RR for ≥ 50% reduction in pain 1.57, 95% CI 1.20 to 2.06; NNTB 8, 95% CI 4 to 21) and over 28 weeks (RR 1.58, 95% CI 1.10 to 2.27) as well as for painful physical symptoms in depression (RR 1.37, 95% CI 1.19 to 1.59; NNTB 8, 95% CI 5 to 14). There was no effect on central neuropathic pain in a single, small, high quality trial. In all conditions, adverse events were common in both treatment and placebo arms but more common in the treatment arm, with a dose‐dependent effect. Most adverse effects were minor, but 12.6% of participants stopped the drug due to adverse effects. Serious adverse events were rare.

Authors' conclusions

There is adequate amounts of moderate quality evidence from eight studies performed by the manufacturers of duloxetine that doses of 60 mg and 120 mg daily are efficacious for treating pain in diabetic peripheral neuropathy but lower daily doses are not. Further trials are not required. In fibromyalgia, there is lower quality evidence that duloxetine is effective at similar doses to those used in diabetic peripheral neuropathy and with a similar magnitude of effect. The effect in fibromyalgia may be achieved through a greater improvement in mental symptoms than in somatic physical pain. There is low to moderate quality evidence that pain relief is also achieved in pain associated with depressive symptoms, but the NNTB of 8 in fibromyalgia and depression is not an indication of substantial efficacy. More trials (preferably independent investigator led studies) in these indications are required to reach an optimal information size to make convincing determinations of efficacy.

Minor side effects are common and more common with duloxetine 60 mg and particularly with 120 mg daily, than 20 mg daily, but serious side effects are rare.

Improved direct comparisons of duloxetine with other antidepressants and with other drugs, such as pregabalin, that have already been shown to be efficacious in neuropathic pain would be appropriate. Unbiased economic comparisons would further help decision making, but no high quality study includes economic data.

PICO

Population
Intervention
Comparison
Outcome

El uso y la enseñanza del modelo PICO están muy extendidos en el ámbito de la atención sanitaria basada en la evidencia para formular preguntas y estrategias de búsqueda y para caracterizar estudios o metanálisis clínicos. PICO son las siglas en inglés de cuatro posibles componentes de una pregunta de investigación: paciente, población o problema; intervención; comparación; desenlace (outcome).

Para saber más sobre el uso del modelo PICO, puede consultar el Manual Cochrane.

Plain language summary

Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia

Review question

Does duloxetine work to treat pain generated by nerves when they have been damaged in disease, or the pain caused by fibromyalgia?

Background

Duloxetine is a drug used to treat depression and urinary urge incontinence (leakage of urine) and it can be also be useful for certain types of pain. Pain can arise spontaneously when there is damage to nerves that carry pain information to the brain (neuropathic pain). When this damage is to nerves outside the spinal cord it is called a peripheral neuropathy. Another type of pain, nociceptive pain, occurs when the nerves sense damage to another tissue (for example, a pinprick in the skin). Some pain is of unclear origin and occurs without apparent nerve or tissue damage. This sort of pain happens, for example, in fibromyalgia. The objective of this review was to assess the benefits and harms of duloxetine for treating painful neuropathy and chronic pain of all sorts.

Study characteristics

We looked at all the published scientific literature and found 18 trials, involving a total of 6407 participants, that were of sufficient quality to include in this review. Eight trials tested the effect of duloxetine on painful diabetic neuropathy and six on the pain of fibromyalgia. Three trials treated painful physical symptoms associated with depression and one small study investigated duloxetine for the pain from strokes or diseases of the spinal cord (central pain).

Key results and quality of the evidence

The usual dose of duloxetine is 60 mg. At this dose, there was moderate quality evidence that duloxetine reduced pain in both painful diabetic peripheral neuropathy and fibromyalgia. In diabetic peripheral neuropathic pain, a 50% or better improvement with duloxetine 60 mg per day was just over one and a half times more likely than with placebo. Another way of saying this is that five people with painful diabetic peripheral neuropathy had to receive duloxetine to achieve a 50% or better response in one person. The effect on fibromyalgia was similar but the number needed to treat for one person to improve by 50% or more was eight. On the basis of a single study it is not possible to determine if a dose of 20 mg is effective, and 120 mg was no more effective than 60 mg.

We calculated that for diabetic neuropathy there have been enough trials to draw these conclusions and no more trials are needed. In fibromyalgia and the painful symptoms associated with depression, more trials are required to make convincing statements about the effectiveness of duloxetine.

Most people taking duloxetine will have at least one side effect. These are mostly minor and the most common are feeling sick, being too awake or too sleepy, headache, dry mouth, constipation or dizziness. About one in six people stop duloxetine because of side effects. Serious problems caused by duloxetine are very rare.

Although duloxetine is beneficial in the treatment of neuropathic pain and fibromyalgia there is little evidence from trials comparing duloxetine to other antidepressant drugs as to which is better.

We have concluded that duloxetine is useful for treating pain caused by diabetic neuropathy and probably fibromyalgia.

The information in this review is up to date to November 2013, the most recent search of the literature.

Authors' conclusions

Implications for practice

There is moderate quality evidence from four studies performed by the manufacturers of duloxetine that doses of 60 mg and 120 mg daily are efficacious for treating pain in diabetic peripheral neuropathy but lower daily doses are not. In fibromyalgia, there is low to moderate quality evidence that duloxetine is effective at similar doses and with a similar magnitude of effect. That effect may be achieved through a greater improvement in mental symptomatology than somatic physical pain. There is also low to moderate quality evidence that pain relief is also achieved in pain associated with depressive symptoms but the NNTB of 8 in fibromyalgia and depression may not make this agent a first line choice if other more efficacious agents are available.

Minor adverse effects are common with duloxetine. They are more common with duloxetine 60 mg than placebo and certainly more common at doses of 120 mg daily. Adverse events were much less frequent at duloxetine 20 mg daily. Serious side effects are rare.

Implications for research

Trial sequential analysis indicates that for painful diabetic neuropathy no further trials are needed to indicate the efficacy of duloxetine at 60 mg. More studies are required to convincingly demonstrate the efficacy of 60 mg of duloxetine in fibromyalgia, the painful physical symptoms of pain in depressive illness and in central pain. These trials should preferably be investigator led and independent of the company making the drug, as bias questions are always raised when the majority or all of the trials are drug company run or sponsored.

The trials were not designed to investigate mechanisms but there was some evidence that the effect on pain was independent from the effect on depression. Improved direct comparisons of duloxetine with other antidepressants and with other drugs, such as pregabalin, already shown to be efficacious in neuropathic pain would be appropriate. All trials should include unbiased economic analyses, although the economic analyses so far performed (including unbiased independent analysis) indicate that duloxetine is cost effective for treating neuropathic pain when tested in a number of models, at least in the US healthcare system.

Summary of findings

Open in table viewer
Summary of findings for the main comparison. Duloxetine for the treatment of painful diabetic neuropathy

Duloxetine for painful diabetic neuropathy

Patient or population: patients with painful neuropathy or chronic pain from diabetic peripheral neuropathy
Settings: primary and secondary care
Intervention: duloxetine

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Duloxetine

Number of patients with ≥ 50% improvement of pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert score

Follow‐up: 8 to 12 weeks

257 per 1000

445 per 1000
(370 to 535)

RR 1.73
(1.44 to 2.08)

908
(4 studies)

⊕⊕⊕⊝
moderate1

NNTB for ≥ 50% reduction in pain at 60 mg daily: 5 (95% CI 4 to 7)

Mean improvement in pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert score

Scale from: 0 to 10
Follow‐up: 8 to 12 weeks

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 60 mg daily in the control groups was
‐1.65 units

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 60 mg daily in the intervention groups was
0.96 lower
(1.26 to 0.65 lower)

722
(4 studies)

⊕⊕⊕⊝
moderate2

Number of patients with ≥ 30% improvement in pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert scale

Follow‐up: 8 to 12 weeks

411 per 1000

629 per 1000
(547 to 719)

RR 1.53
(1.33 to 1.75)

799
(4 studies)

⊕⊕⊕⊝
moderate1

NNTB for ≥ 30% reduction in pain at 60 mg duloxetine daily: 5 (95% CI 3 to 7)

Mean improvement in Patient Reported Global Impression of Change at 12 weeks or less

Duloxetine 60 mg daily
VAS

Scale from: 0 to 10
Follow‐up: 8 to 12 weeks

The mean mean improvement in patient reported global impression of improvement change at 12 weeks or less ‐ duloxetine 60 mg daily in the control groups was
‐3.06 units

The mean mean improvement in Patient Reported Global Impression of Improvement Change at 12 weeks or less ‐ duloxetine 60 mg daily in the intervention groups was
0.6 lower
(0.77 to 0.44 lower)

1018
(5 studies)

⊕⊕⊕⊝
moderate3

Adverse event leading to cessation

All neuropathic pain indications

Duloxetine 60 mg daily

56 per 1000

109 per 1000
(90 to 133)

RR 1.95
(1.6 to 2.37)

4837
(14 studies)

⊕⊕⊝⊝
low4

NNTH for duloxetine 60 mg daily, all indications, and all adverse effects leading to cessation: 18 (95% CI 13 to 30)

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; NNTB: number needed to treat for an additional beneficial outcome; NNTH: number needed to treat for an additional harmful outcome

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

1 Four trials, all company sponsored and performed but all trials pre‐registered on ClinicalTrials.gov have been published. No publication bias detected.
2 Two of four studies by company. Effect in Rowbotham nonsignificant, contributing some heterogeneity.
3 Five studies but wide CIs in the independent studies.
4 Variable quality of adverse event collection.

Open in table viewer
Summary of findings 2. Duloxetine for the treatment of the chronic pain of fibromyalgia

Duloxetine for the chronic pain of fibromyalgia

Patient or population: patients with the chronic pain of fibromyalgia
Settings:
Intervention: duloxetine

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Duloxetine

Number with ≥ 50% improvement of pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert scale
Follow‐up: 8 to 12 weeks

233 per 1000

366 per 1000
(280 to 480)

RR 1.57
(1.2 to 2.06)

528
(2 studies)

⊕⊕⊝⊝
low1,2

NNTB for ≥ 50% improvement of pain at duloxetine 60 mg daily: 8 (95% CI 4 to 21)

Number with ≥ 30% improvement of pain at 12 weeks or less

Duloxetine 60 mg daily

Follow‐up: 8 to 12 weeks

347 per 1000

527 per 1000
(430 to 642)

RR 1.52
(1.24 to 1.85)

528
(2 studies)

⊕⊕⊝⊝
low1,2

NNTB for ≥ 30% improvement of pain at duloxetine 60 mg daily: NNT 6 (95% CI 3 to 12)

Mean improvement in the Patient Reported Global Impression of Change at completion of trial

Duloxetine 60 mg daily
VAS

Scale from: 0 to 10
Follow‐up: 12 weeks

The mean mean improvement in the patient reported global impression of change at completion of trial ‐ duloxetine 60 mg daily in the control groups was
3.52 units

The mean mean improvement in the patient reported global impression of change at completion of trial ‐ duloxetine 60 mg daily in the intervention groups was
0.45 lower
(0.73 to 0.18 lower)

519
(2 studies)

⊕⊕⊝⊝
low1,2

Mean improvement in pain at 12 weeks or less

Duloxetine 120 mg daily

LikertScale from: 0 to 10
Follow‐up: 12 weeks

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 120 mg daily in the control groups was
‐1.5

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 120 mg daily in the intervention groups was
0.8 lower
(1.35 to 0.25 lower)

507
(1 study)

⊕⊕⊕⊝
moderate1

Adverse events

See comment

See comment

See comment

See comment

See pooled adverse events in 'Summary of findings' table 1

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; NNTB: number needed to treat for an additional beneficial outcome

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

1 Substantial dropouts from all trials inform the outcomes.
2 Mostly female in some trials, all female in others.

Open in table viewer
Summary of findings 3. Duloxetine for the treatment of pain in major depressive disorder

Duloxetine for pain in major depressive disorder

Patient or population: patients with pain in major depressive disorder
Settings:
Intervention: duloxetine

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Duloxetine

Number with ≥ 50% pain relief at 12 weeks or less
Follow‐up: 12 weeks

360 per 1000

493 per 1000
(428 to 572)

RR 1.37
(1.19 to 1.59)

1023
(2 studies)

⊕⊕⊕⊝
moderate1

NNTB for ≥ 50% pain relief at < 12 weeks 60 mg duloxetine daily: 8 (95% CI 5 to 14)

Number with ≥ 30% pain relief at 12 weeks or less

467 per 1000

593 per 1000
(537 to 654)

RR 1.27
(1.15 to 1.4)

1359
(3 studies)

⊕⊕⊝⊝
low1,2

NNTB for ≥ 30% pain relief at < 12 weeks 60 mg duloxetine: 8 (95% CI 4‐ to 14)

Mean improvement in pain at 12 weeks or less
Visual analogue scale. Scale from: 0 to 10.
Follow‐up: 12 weeks

The mean mean improvement in pain at 12 weeks or less in the control groups was
1.23

The mean mean improvement in pain at 12 weeks or less in the intervention groups was
0.55 lower
(0.75 to 0.35 lower)

1359
(3 studies)

⊕⊕⊝⊝
low1,2

Mean improvement in Patient Reported Global Impression of Change at 12 weeks or less

See comment

See comment

Not estimable

See comment

Outcome not measured

Adverse events

See comment

See comment

Not estimable

See comment

See pooled adverse events in 'Summary of findings' table 1

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; NNTB: number needed to treat for an additional beneficial outcome

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

1 Mixed causes for pain, not necessarily neuropathic.
2 Substantial dropouts partially accounted for by last observation carried forward and statistical manipulation.

Background

Description of the condition

Pain is common in peripheral nerve diseases such as the peripheral neuropathy associated with diabetes mellitus. Painful neuropathy is a particular example of neuropathic pain. Neuropathic pain is "pain arising as a direct consequence of a lesion or disease affecting the somatosensory system" (Treede 2008). Neuropathic pain is different from conventional or nociceptive pain. Nociceptive pain arises from the activation of primary pain receptors in response to injury or inflammation. Painful neuropathies are diseases of the peripheral nerves that cause neuropathic pain.

Chronic pain has been classified as pain exceeding three months' duration (Nagda 2004). Chronic pain is a major health problem affecting one in five people in Europe (Breivik 2006). A community based study in North West England estimated the prevalence of chronic painful peripheral neuropathy in people without diabetes as 4.9% (Daousi 2004).The prevalence in people with diabetes in the same community was 16.2%. In a large community study in the United Kingdom, the annual incidence of neuropathic pain was calculated as at least 84 per 100,000 by adding the incidence of the four commonest and most disabling causes (diabetic neuropathy, trigeminal neuralgia, postherpetic neuralgia and phantom limb pain) (Hall 2006).

The pain of painful peripheral neuropathy can be diverse and distressing. Descriptions include burning, cold, electric shocks, lancinating, tight or aching. Other spontaneous and evoked positive sensory symptoms include painful numbness, tingling or paraesthesiae. Stimuli that are not usually painful may be perceived as painful, a phenomenon called allodynia. Chronic pain can have serious complex adverse psychological and social effects.

Description of the intervention

Duloxetine is one of a newer type of antidepressant drug. It is a relatively balanced dual reuptake inhibitor of serotonin and noradrenaline (Schuessler 2006). Theoretically these actions should make it a good pain modulating agent (Bymaster 2001; Bymaster 2005). Serotonin modulates both pro‐nociceptive and anti‐nociceptive descending effects on central pain pathways from the brainstem. Noradrenaline has a predominantly anti‐nociceptive effect. Balance between facilitation and depression of pain pathways is important for normal function. Drugs that inhibit the reuptake of serotonin and noradrenaline potentiate monoamine neurotransmission in the descending inhibitory spinal pathways and so reduce nociceptive afferent transmission in the ascending spinal pain pathways. Potentiation of both serotonin and noradrenaline is required to produce effective analgesia. The action of drugs such as duloxetine is independent of their effects on depression (Perahia 2006). Onset of benefit occurs within days, earlier and at lower doses than in depression. Furthermore, they have similar effects on pain in depressed and non‐depressed people. Common side effects include nausea, headache, dry mouth, insomnia, constipation, dizziness, fatigue, somnolence, hyperhydrosis and diarrhoea (Gahimer 2007). These are mainly classified as mild to moderate and anecdotally appear less prevalent than the side effects with tricyclic antidepressants.

Why it is important to do this review

Duloxetine is licensed in the United States, European Union and United Kingdom for the treatment of major depressive disorder (Nose 2007), urinary stress incontinence (Mariappan 2009), and for the management of neuropathic pain associated with diabetic peripheral neuropathy. We do not know of a published systematic review of duloxetine for any pain condition and it was not included in a previous Cochrane review of antidepressants for neuropathic pain (Saarto 2007). This review aims to fill the gap. Painful neuropathy is the principal focus of this review because duloxetine has been chiefly tried for this indication. However, previous Cochrane reviews of interventions for pain have covered all forms of either neuropathic pain or acute and chronic pain. For conformity with these other reviews, we will include all forms of chronic pain which have a neuropathic component, chronic pain with no explanation and fibromyalgia, but not acute pain, for which duloxetine has not been proposed as a treatment, or pain from specific non‐neuropathic causes covered in other reviews (for example pain from osteoarthritis of the knee).

Objectives

To assess the benefits and harms of duloxetine for treating painful neuropathy and different types of chronic pain.

Methods

Criteria for considering studies for this review

Types of studies

For the detection of benefits, we included only double‐blind randomised trials of duloxetine for treating painful neuropathy or chronic neuropathic pain, chronic pain conditions without identified cause or fibromyalgia. Duloxetine was to have been administered for a minimum of eight weeks. We included eligible studies irrespective of publication status or language of publication.

Types of participants

We included participants with any form of painful peripheral neuropathy, chronic neuropathic pain, chronic pain conditions without identified cause, or fibromyalgia.

Types of interventions

We included all formulations and doses of duloxetine in comparison with placebo or other controls. We reported comparisons with placebo and with other controls separately.

Types of outcome measures

Primary outcomes

The primary outcome was short‐term (up to and including 12 weeks) improvement of pain compared with baseline using validated scales of pain intensity or pain relief. We accepted both visual analogue and categorical scales. Where reports expressed pain relief as none, minor, moderate, major or complete, we considered only moderate, major or complete as improvement. Where studies measured pain with a continuous scale, we took improvement to be an improvement of 50% or more from baseline on that scale. If studies reported results only as improvement on a continuous scale, we planned to try to obtain results from the authors to provide this dichotomous analysis.

Secondary outcomes

  1. Long‐term (more than 12 weeks) improvement of pain compared with baseline, analysed as for the primary outcome.

  2. Improvement in short‐term (up to and including 12 weeks) and long‐term (more than 12 weeks) pain of at least 30% compared with baseline using validated scales of pain intensity and or pain relief, analysed as for the primary outcome.

  3. Improvement in any validated quality of life score of 30% or more compared to the baseline.

  4. As the outcome measures for the assessment of pain were likely to be diverse and the majority of trials use standard subjective scales for pain intensity or pain relief or both, further results were to be analysed according to the third to sixth types in a hierarchy modified from Wiffen 2005. The full hierarchy of outcome measures is as follows.

    1. Patient reported pain relief of 50% or greater.

    2. Patient reported pain relief of 30% or greater.

    3. Patient reported global impression of clinical change (PGIC).

    4. Pain on movement.

    5. Pain on rest.

    6. Any other pain related measure.

  5. Adverse events during treatment. We analysed categories of: all adverse events, severe or serious adverse events that led to hospitalisation or death, and adverse events leading to cessation of treatment.

We chose 30% and 50% as the percentage of pain improvement considered clinically important for dichotomous outcomes, in line with the recommendations made by the Initiative on Methods, Measurement, and Pain Assessment in Clinical Trials (IMMPACT) (Dworkin 2008). Improvement in pain intensity of 30% or more is considered moderately important and 50% or more, substantial improvement.

Outcomes for inclusion in a 'Summary of findings' table

We created a 'Summary of findings' table for each included neuropathic pain condition for which meta‐analysis was possible, using the following outcomes.

  1. Number of participants with ≥ 50% improvement of pain at 12 weeks or less.

  2. Mean improvement in pain at 12 weeks or less.

  3. Number of participants with ≥ 30% improvement in pain at 12 weeks or less.

  4. Mean improvement in PGIC at 12 weeks or less.

  5. Adverse event leading to cessation.

We used the five GRADE considerations (study limitations, consistency of effect, imprecision, indirectness and publication bias) to assess the quality of a body of evidence (studies that contribute data for the prespecified outcomes). We used methods and recommendations described in Section 8.5 and Chapter 12 of the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011) using GRADEpro software (GRADEpro 2008). We justified decisions to down‐ or up‐grade the quality of studies using footnotes.

Search methods for identification of studies

Electronic searches

We searched the specialised registers of the Cochrane Neuromuscular Disease Group and the Cochrane Pain, Palliative and Supportive Care Group (PaPaS), CENTRAL (2013, Issue 11), MEDLINE (January 1966 to November 2013) and EMBASE (January 1980 to November 2013). We searched the National Institutes for Health clinical trials registry ClinicalTrials.gov (www.clinicaltrials.gov) up to April 2013 for current ongoing registered trials. We also searched DARE (Database of Abstracts of Reviews of Effects), HTA (Health Technology Assessment) and NHSEED (NHS Economic Evaluation Database) (2013, Issue 4 in The Cochrane Library), for papers for inclusion in the Discussion. The detailed search strategies are in the appendices: MEDLINE (Appendix 1), EMBASE (Appendix 2), CENTRAL (Appendix 3), Cochrane Neuromuscular Disease Group Specialized Register Appendix 4 and ClinicalTrials.gov Appendix 5.

Searching other resources

We also wrote to Eli Lilly who make duloxetine and to pain experts asking for information about other or ongoing trials. We searched the Lilly online trials database (http://www.lillytrials.com/) for other trials not identified in the above searches. We wrote to the authors of studies to clarify aspects of trial design that were unclear from the published papers.

Data collection and analysis

Selection of studies

Two review authors (MLand RACH) independently scrutinised all the titles and abstracts revealed by the searches and determined which trials fulfilled the selection criteria. They resolved disagreement by discussion without the need to involve the third review author (PW).

Data extraction and management

Two review authors (ML and RACH) extracted data independently onto a specially designed data extraction form. We would have resolved disagreements by discussion if necessary with the third review author (PW) but this was not necessary. One author (ML) entered data into the Cochrane software, Review Manager 5 (RevMan), and a second author (RACH) checked them.

Assessment of risk of bias in included studies

We used the methods of the Cochrane Collaboration to assess 'Risk of bias' as set out in Chapter 8 of the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2008, updated Higgins 2011) rather than those stipulated in the protocol for this review, which predated the new methods.

Measures of treatment effect

The effect measures of choice were the risk ratio (RR) for dichotomous data and the mean difference (MD) or standardised mean difference (SMD) for continuous data. We expressed uncertainty with 95% confidence intervals (CIs). We also expressed the most important results as numbers needed to treat for an additional beneficial outcome (NNTB) and numbers needed to treat for an additional harmful outcome (NNTH), where appropriate.

Unit of analysis issues

Cross‐over trials

We would have analysed cross‐over trials using the estimated differences in effects and their standard errors with the generic inverse variance (GIV) facility in RevMan if we had the necessary data. For results using dichotomous outcomes this would have been more difficult, but we would have used this approach if we could have converted the results to odds ratios (OR) on the log scale and calculated the standard errors. If necessary we would also have analysed the results following the methods of Elbourne 2002 with the assistance of a statistician.

Assessment of reporting biases

If there had been sufficient trials, we would have inspected funnel plots for asymmetry that might have been due to publication bias. We were aware that funnel plots and statistical tests based on them are not reliable indicators of publication bias and we would have treated any interpretations made from them with great caution.

Data synthesis

We undertook each meta‐analysis using a fixed‐effect model in the RevMan software. We used the I2 statistic for heterogeneity and if its value had been greater than 50% we would have inspected the trials, forest plots and L'Abbé plots for differences between trials that might have explained the heterogeneity. In the absence of any explanation, we would have repeated the analysis with a random‐effects model.

Subgroup analysis and investigation of heterogeneity

We reported results for painful diabetic neuropathy, fibromyalgia, chronic pain and non‐specific bodily pain associated with depression separately, and would have reported other specific causes of neuropathic pain, namely trigeminal neuralgia, postherpetic neuralgia and central ('thalamic') pain separately. In addition to reporting the results of all forms of painful neuropathy together, we would also have reported the results for the following different diagnostic subgroups separately: diabetic neuropathy, HIV neuropathy, and idiopathic painful neuropathy.

Sensitivity analysis

We intended to conduct the following sensitivity analyses:

  1. trials that did and did not have perfect scores for 'Risk of bias';

  2. trials with more than or less than 20% dropout or loss to follow‐up;

  3. trials that were and were not led by the company producing the drug; and

  4. trials with more than and fewer than 100 participants.

A sensitivity analysis was possible only in the trials of diabetic neuropathy and only in the context of studies with a less than a 20% dropout rate.

Trial sequential analysis

We performed trial sequential analysis (TSA) using software provided by the Copenhagen Trial Unit (Thorlund 2011). We used predefined limits to frame the statistical analysis, with a conventional analysis. Limits were alpha 0.05, beta 0.1, relative risk 0.66, and we defined the placebo rate according to that found in the extracted meta‐analysis data. We performed TSA on the primary outcome for each of the conditions included in the review (diabetic neuropathy, fibromyalgia, central pain, and painful physical symptoms in depression).

Adverse events

Randomised trials may not capture all important adverse events, but this systematic review now contains data from more than 6000 participants and the adverse effects reported were fairly consistent across all studies. It is noted that the drug manufacturer conducted all but one of the studies.

Economic issues

We considered costs in the Discussion.

We reported any changes from the published protocol of the review (Lunn 2008) in Differences between protocol and review.

Results

Description of studies

Results of the search

In 2009, for the original review, we identified 130 references to possible trials (MEDLINE 12, EMBASE 75, CENTRAL 19, Cochrane Neuromuscular Disease Group Specialized Register and PaPaS Group Register and Library 22, and handsearches 2). Following exclusion of duplicates and studies that were clearly irrelevant, two authors checked 37 titles and identified 14 RCTs or possible RCTs. From these we selected six trials for inclusion. In 2012 to 2013, we performed a database search extension to October 2012 and updated the search of www.clinicaltrials.gov to April 2013. The search for RCTsretrieved 298 new references (MEDLINE 120, EMBASE 156, CENTRAL 21, and the specialised registers of the Cochrane Neuromuscular Disease Group and PaPaS 1). We performed another search to November 2013 and identified a further 150 references including references to economic analyses and a number of other systematic reviews.

Two authors selected potentially eligible references from this list and after deduplication, 47 possible references remained. We found a further study from review of the reference lists of identified papers and another study from querying publications from studies ongoing in the first version of this review and now published. After discussion of titles and abstracts, we selected 27 new references for full‐text review, of which we included 12. There are therefore 18 trials in total in this update.

Included studies

The 18 studies in this Cochrane Systematic Review include a total of 6407 participants, and cover painful neuropathy, chronic neuropathic pain (in this review central pain from strokes or spinal cord disorders), fibromyalgia and painful physical symptoms (of unknown cause) in depressive disorders. We excluded trials of duloxetine in conditions where pain is from another disease where the pain is not neuropathic (for example, we excluded osteoarthritis and pelvic pain), but included neuropathic pain diagnoses associated with specific neural injury such as spinal cord injuries, multiple sclerosis, or stroke. The manufacturers of duloxetine, Eli Lilly, were the sponsors of all but one of the included studies (Vranken 2011).

We described the characteristics of the classified and included studies in Characteristics of included studies. Eight studies, including 2728 participants, looked at duloxetine in the treatment of painful diabetic peripheral neuropathy.

Six studies, involving 2249 participants, tested duloxetine for fibromyalgia. Four studies tested duloxetine for 12 weeks (Arnold 2004; Arnold 2005; Arnold 2010; Arnold 2012) and two for six months (Russell 2008; Chappell 2008).

Three studies (1382 participants) examined the effect of duloxetine in participants who had painful physical symptoms unexplained by any known alternative diagnosis in the context of major depressive disorder (Brecht 2007; Gaynor 2011a; Gaynor 2011b).

One study (Vranken 2011) examined the effect of duloxetine in 48 participants with central neuropathic pain.

Six of the eight studies in diabetic neuropathy compared duloxetine with placebo in parallel groups for two to three months (Goldstein 2005; Raskin 2005; Wernicke 2006; Gao 2010; Yasuda 2010; Rowbotham 2012). One study compared duloxetine to amitriptyline in a cross‐over design with only six weeks' treatment in each arm with a short two‐week washout (Kaur 2011), but because it is the only comparative trial of its type, we included some discussion of it but have not included it in meta‐analysis. Kaur 2011 and Gao 2010 had variable dosage schedules for duloxetine (Kaur 20 mg to 60 mg and Gao 60 mg to 120 mg). We carried out analyses for benefit as if all participants were on the higher dose and for harms as if all were on the lower dose. One study compared duloxetine to pregabalin in the randomised parallel group first arm of an enrichment trial design (Tesfaye 2013), but we used only the randomised parallel group study period II prior to enrichment in the meta‐analysis as it was unclear whether there was any rerandomisation in Study Period III.

Five of the included studies of diabetic peripheral neuropathy were broadly similar in design and were all conducted by the same drug company (Goldstein 2005; Raskin 2005; Wernicke 2006; Gao 2010; Yasuda 2010). Participants all had to be at least 18 years old, to have had a length‐dependent painful peripheral neuropathy caused by either type I or type II diabetes for at least six months, and had to have a diagnosis of diabetes on a validated published scale and a reasonable minimum average 24‐hour pain score (for example 4 on an 11‐point Likert scale or > 50% on a VAS of pain). The age, sex, pain severity and duration of pain at entry for the participants were similar in the treatment groups in each trial and between trials, except Yasuda 2010, in which three‐quarters of the participants were male. In these five trials participants were treated with duloxetine in oral capsule or tablet form. Doses varied between trials: in Yasuda 2010, participants were treated with doses of 40 mg or 60 mg; in Goldstein 2005, Raskin 2005 and Wernicke 2006, dosage was 60 mg once or twice per day or identical placebo, with the addition of a 20 mg once daily dose in the trial of Goldstein. Gao 2010 commenced with 60 mg duloxetine but this could increase to 120 mg after two weeks if the participant had an inadequate response. Treatment was for 12 weeks with a one week taper in four trials (Raskin 2005; Wernicke 2006; Gao 2010; Yasuda 2010). All five trials took place in healthcare and research centre settings. Tesfaye 2013 compared pregabalin or duloxetine at doses increasing to their maximum with a combination of the two drugs together. The seventh trial was of an α4ß2 neuronal nicotinic receptor agonist ABT‐894 and compared the efficacy of ABT‐894 to that of placebo and to duloxetine 60 mg over eight weeks (Rowbotham 2012).

The six studies of fibromyalgia included participants aged 18 or older who fulfilled American College of Rheumatology criteria for fibromyalgia. Five of the six studies stipulated minimum entry criteria: participants had significant pain at entry (≥ 4 on the pain intensity item of the Fibromyalgia Impact Questionnaire (Arnold 2004) or Brief Pain Inventory (Arnold 2005; Russell 2008; Arnold 2010; Arnold 2012)). One study (Chappell 2008) did not stipulate any criteria for pain at entry and participants could have, or not have, major depressive disorder. These trials were also conducted by the same drug company that performed the diabetic neuropathy studies. One study included only women (Arnold 2005) and the other five included over 90% women, despite being open to males and females, reflecting the epidemiology of this condition. In the five studies that gave ages (Arnold 2004; Chappell 2008; Russell 2008; Arnold 2010; Arnold 2012), the participants were approximately 10 years younger than in the diabetic peripheral neuropathy trials. Participants had similar levels of pain at entry in each trial even though there were no pain entry criteria for Chappell 2008. Only Arnold 2005 stated the duration of pain at entry (> 12 weeks). All six trials were blinded. Participants in Arnold 2004, Arnold 2005 and Russell 2008 received duloxetine in capsules or identical appearing placebos in identical dosage schedules in outpatient research facilities for 12 weeks (Arnold 2004; Arnold 2005), or for 28 weeks (Russell 2008). Arnold 2010 and Chappell 2008 used tablets and also a rather more complex dosing schedule, where the starting dose was 60 mg once daily with a 30 mg run‐in phase for one week. In Chappell 2008, there was then a randomised increase to 120 mg after 13 weeks if participants had not reached a reduction of > 50% in pain on the BPI average pain score. In Arnold 2010, after a one‐week 30 mg run‐in, all participants in the active arm received 60 mg; the treating physician then increased the dose after four weeks to 90 mg or 120 mg if there was less than 50% improvement on the BPI scale (participants were blinded to the increase in dose). The final doses at week 12 were: 60 mg, n = 137 (52.1%); 90 mg, n = 62 (23.6%); and 120 mg, n = 64 (24.3%). The Russell 2008 trial included a two‐week titration phase and a two‐week taper. Finally, Arnold 2012 used a low dose of 30 mg only, presented as capsules.

We included three studies of painful physical symptoms in major depressive disorder (MDD) (Brecht 2007; Gaynor 2011a; Gaynor 2011b). The trials included participants if they had a diagnosis of MDD and painful physical symptoms and were devoid of an alternative pain syndrome. Treatment in all three studies was eight weeks of duloxetine 60 mg or placebo and although there was a slightly higher proportion of men in the studies of Gaynor et al. than in Brecht 2007, the studies were otherwise well matched for pain, age, depression scores and other demographic characteristics. The studies provide no details of the quality or somatic distribution of the types of pain experienced.

One study (Vranken 2011) examined the effect of duloxetine compared versus placebo in participants with central neuropathic pain. Those eligible were over 18 years old with more than six months' severe neuropathic pain of spinal cord or cerebrovascular origin. Participants had a score of more than six on a 10‐point VAS. This trial was not company‐sponsored. The starting dose of duloxetine was initially 60 mg, which increased if participants did not improve by more than 1.8 points on VAS.

Excluded studies

We excluded 29 trials for a number of reasons (see Characteristics of excluded studies). Two excluded texts were summary reports of other studies that we have included (Raskin 2005a; Russell 2006). Of the trials that did not meet the pre‐defined criteria for inclusion in this review, seven were for conditions outside the remit of the review (NCT01451606; Chappell 2009; Skljarevski 2008; Skljarevski 2009; Skljarevski 2010; Skljarevski 2010b; Chappell 2011). Eight were open studies without blinding or a control group (NCT00125892; NCT00385671; NCT00552682; NCT00641719; Raskin 2006b; Skljarevski 2009a; Tanenberg 2011; Wu 2006). Canovas 2007 was neither randomised nor controlled. The Raskin 2006a trial (in diabetic peripheral neuropathic pain) was randomised but not blinded; it is mentioned in the Discussion of this review. The Wernicke 2006b study was a 52‐week extension of the Goldstein 2005 randomised trial with a similar design but without blinding. The NCT00266643, Brannan 2005, Vollmer 2011, Boyle 2012, Lavoie Smith 2012, Smith 2013 (abstract of Lavoie Smith 2012) and Harrison 2013 trials measured outcomes at durations of less than eight weeks, not the eight weeks stipulated in our protocol. Goldstein 2004 tested duloxetine for eight weeks for depression, but included pain scales as secondary outcome measures. However, it was not clear what sort of pain the participants had (for example musculoskeletal, neuropathic, or headache) and the levels of pain at baseline were low compared to the included trials. NCT00425230 was registered in ClinicalTrials.gov but was terminated before inclusion of participants.

Ongoing and completed but unpublished trials

A search of ClinicalTrials.gov revealed five ongoing studies in pain from multiple sclerosis (1 study), diabetic neuropathy (2 studies) and fibromyalgia (2 studies) (NCT00457730; NCT00619983; NCT01179672; NCT01237587; NCT01552057) (see Characteristics of ongoing studies). More importantly, we were unable to find results for five potentially eligible studies registered as 'completed' in ClinicalTrials.gov (NCT00125892, NCT00233025, NCT00489073, NCT00603265 and NCT01579279), as the results were not available in ClinicalTrials.gov, the trials were not identifiable as publications in the Lilly Trials register or a publication database, and we were unable to obtain the study reports through writing to the study investigators, where identifiable. Vollmer 2011 may be NCT00755807 on ClinicalTrials.gov, where results are presented, but although there is a published abstract there is no published full paper at the time of publication of this review.

Risk of bias in included studies

The 18 included studies were variable in their risk of bias (see Figure 1 and Characteristics of included studies). Eight of the studies were at a high risk of bias for at least one attribute. Seven of the studies had an unclear risk of bias in one or more domains, because of various problems. In only three studies was the risk of bias deemed to be low across all the attributes; one of the three being a study from the drug company. Interestingly, despite common authors across studies and development by the company of methodology and outcomes selection over time, the risk of bias in studies did not improve and some data are noticeably absent from later studies. Nearly all the studies had a dropout rate of more than 20% (only Raskin 2005, Gao 2010, Yasuda 2010 and Tesfaye 2013 had a dropout rate of less than 20%), which was deemed to change the overall risk of bias from low to unclear. Dropouts in Rowbotham 2012 (a relatively small study of the novel Abbott agent ABT‐894 versus duloxetine) and Arnold 2012 (a negative trial that used a subtherapeutic dose of duloxetine) were particularly low. The same company sponsored and performed all but one of the included studies (Vranken 2011).


Methodological quality summary: review authors' judgements about each methodological quality item for each included study. Green = low risk of bias; yellow = unclear risk of bias; red = high risk of bias

Methodological quality summary: review authors' judgements about each methodological quality item for each included study. Green = low risk of bias; yellow = unclear risk of bias; red = high risk of bias

The issue of incomplete outcome data in Arnold 2004 was unclear from the publication; 36% of the placebo group and 44% of the duloxetine group discontinued the study. From information provided by the authors, the analysis included all participants with at least one follow‐up measurement from baseline, with the last observation carried forward.

A number of trials used exploratory assessments of statistical processing including presentation of multiple statistical analyses, for example last observation carried forward (LOCF) and baseline observation carried forward (BOCF) data, leading to a suspicion of post hoc data mining using exploratory statistics.

Effects of interventions

See: Summary of findings for the main comparison Duloxetine for the treatment of painful diabetic neuropathy; Summary of findings 2 Duloxetine for the treatment of the chronic pain of fibromyalgia; Summary of findings 3 Duloxetine for the treatment of pain in major depressive disorder

We analysed the effects of the interventions for painful peripheral neuropathy, fibromyalgia, central pain, and painful physical symptoms in major depressive disorder separately; we did not perform meta‐analysis combining all of the trials across the four 'conditions'. We identified no includable trials of duloxetine for other causes of neuropathic pain, although many low quality studies exist in other diseases such as post herpetic neuralgia.

All studies included adverse event data that had been sought prospectively and which the trial authors reported in detail. We analysed these across conditions.

None of the randomised trials included health economic data.

Painful peripheral neuropathy ‐ duloxetine versus placebo

Primary outcome: short‐term (up to and including 12 weeks) improvement of pain compared with baseline

Five trials in painful diabetic neuropathy reported data on the primary outcome measure of ≥ 50% improvement of pain compared with baseline at less than 12 weeks (Goldstein 2005; Raskin 2005; Wernicke 2006; Gao 2010; Yasuda 2010). Participants received duloxetine 20 mg, 40 mg, 60 mg or 120 mg per day. Combining data from all doses from the five trials together (1655 participants), the RR of ≥ 50% improvement with any dose was 1.53 (95% CI, 1.21 to 1.92) compared with placebo (see Figure 2, Analysis 1.1). The RR of improvement was significantly greater than placebo for the 40 mg, 60 mg and 120 mg daily doses but not the 20 mg daily dose, for which it was 1.43 (95% CI 0.98 to 2.09; the CIs for 20 mg were wide as only one study with few participants provided data). There was no significant difference nor a dose effect in the RR of improvement with increasing doses of duloxetine from 40 mg to 120 mg. Significant heterogeneity in the 'all doses' and the 120 mg dose analysis is explained by the inclusion of Gao 2010. As it was not clear how many participants completed that trial on doses of 60 mg or 120 mg daily, we assumed for the purposes of the analysis that the higher dose was reached by all. Removing Gao 2010 removed the heterogeneity and slightly increased the RR of benefit for 'all doses' (RR 1.68, 95% CI 1.41 to 2.02).


Duloxetine versus placebo in the treatment of painful neuropathy: Number of patients with >50% improvement of pain at <12 weeks.

Duloxetine versus placebo in the treatment of painful neuropathy: Number of patients with >50% improvement of pain at <12 weeks.

The mean improvement in pain at 12 weeks or less on an 11‐point Likert scale was significantly greater than placebo with the 60 mg dose of duloxetine (MD ‐0.96, 95% CI ‐1.26 to ‐0.65; 4 trials, 722 participants) and the 120 mg dose (MD ‐0.93, 95% CI ‐1.21 to ‐0.65; 4 trials, 828 participants), but not with the 20 mg dose (see Figure 3, Analysis 1.2, 1 trial, 179 participants, wide CIs from the single study). Removal of the Gao 2010 data removed the heterogeneity contributed by this study and the data then indicated a dose effect (MD ‐1.16, 95% CI ‐1.49 to ‐0.83; 3 trials, 612 participants) (see Analysis 1.2, Figure 3).


Duloxetine versus placebo in the treatment of pain: Mean improvement in pain at 12 weeks.

Duloxetine versus placebo in the treatment of pain: Mean improvement in pain at 12 weeks.

The quality of the evidence available for this outcome remains moderate, mainly as a result of relatively high dropout rates. Lilly sponsored and performed all of these studies but there is no significant suspicion of publication bias despite a number of trials remaining without an identified publication in ClinicalTrials.gov.

Secondary outcomes

None of the included trials of painful diabetic neuropathy reported outcomes at more than 12 weeks.

Five trials included data on ≥ 30% improvement of pain at 12 weeks or less (Raskin 2005; Wernicke 2006; Gao 2010; Yasuda 2010; Rowbotham 2012). The results were similar to those for at least 50% improvement, as was the heterogeneity introduced by Gao 2010. Relative rates of improvement were significantly greater than placebo with duloxetine for the 40 mg dose (RR 1.57, 95% CI 1.18 to 2.07; 1 trial, 252 participants), the 60 mg dose (RR 1.53, 95% CI 1.33 to 1.75; 4 trials, 799 participants), the 120 mg dose (RR 1.38, 95% CI 1.21 to 1.58; 3 trials, 659 participants) and for all three doses combined (RR 1.45, 95% CI 1.30 to 1.63; 4 trials, 1220 participants) (see Figure 4, Analysis 1.3). With Gao excluded for heterogeneity, the RR for 120 mg is 1.55, 95% CI 1.30 to 1.86 (444 participants), and for all doses, 1.57, 95% CI 1.37 to 1.80 (1005 participants). Data for this outcome for the 20 mg dose were not available.


Duloxetine versus placebo in the treatment of pain: Number of patients with >30% improvement in pain at <12 weeks.

Duloxetine versus placebo in the treatment of pain: Number of patients with >30% improvement in pain at <12 weeks.

Trials that included quality of life information used the SF‐36. We included data on the relevant physical, mental and bodily pain subsections of the SF‐36. In painful diabetic neuropathy, the effect of 20 mg duloxetine was not significantly different from placebo on any of the selected SF‐36 subscores at 12 weeks or less (Raskin 2005; Wernicke 2006; Rowbotham 2012), or the mental subscore at 60 mg daily doses. The MD of improvement in the physical summary component was significantly greater than placebo with the 60 mg dose (2.65, 95% CI 1.38 to 3.92; 3 trials, 514 participants) and 120 mg dose (2.80, 95% CI 1.04 to 4.55; 2 trials, 409 participants) (see Analysis 1.4). The MD on the mental summary component was significantly greater than placebo only with the 120 mg dose (2.23, 95% CI 0.69 to 3.77; 2 trials, 409 participants) (see Analysis 1.5). The MD on the bodily pain subscale showed significantly more improvement than placebo with the 60 mg dose (5.58, 95% CI 1.74 to 9.42; 2 trials, 421 participants) and even more with the 120 mg dose (8.19, 95% CI 4.33 to 12.05; 2 trials, 420 participants) but not with the 20 mg dose (1 trial, 209 participants) (see Analysis 1.6).

Six studies reported the PGIC (Gao 2010; Goldstein 2005; Raskin 2005; Rowbotham 2012; Wernicke 2006; Yasuda 2010), three reported pain at rest (night pain) (Goldstein 2005; Raskin 2005; Wernicke 2006), and two reported the Brief Pain Inventory (BPI) (Raskin 2005; Wernicke 2006). Mean improvements only were reported. The MD versus placebo for each outcome was not significant for the 20 mg dose (1 study, 219 participants), but was significant and similar in magnitude for the 60 mg and 120 mg doses (see Figure 5, Analysis 1.7). However, a minimum clinically meaningful difference in the PGIC is suggested as one point (Dworkin 2008), and hence the change associated with 60 mg duloxetine (MD ‐0.60, 95% CI ‐0.77 to ‐0.44; 5 trials, 1018 participants) is unlikely to be clinically significant. The RR for the BPI with duloxetine 60 mg is statistically significantly reduced by ‐0.97 (95% CI ‐1.38 to ‐0.57; 2 trials, 433 participants), which borders on the change considered clinically significant (Dworkin 2008) (see Analysis 1.8; Figure 6). With duloxetine 120 mg, the MD reached the minimum clinically significant threshold (‐1.16, 95% CI ‐1.91 to ‐0.41; 2 trials, 428 participants). The mean difference of improvement in pain at rest at 12 weeks was significantly greater than placebo with duloxetine 60 mg and 120 mg daily (2 trials, 664 participants), but not with 20 mg daily (1 trial, 222 participants) (Analysis 1.9).


Duloxetine versus placebo in the treatment of pain: Patient reported global impression of change.

Duloxetine versus placebo in the treatment of pain: Patient reported global impression of change.


Duloxetine versus placebo in the treatment of pain: BPI severity ‐ average pain.

Duloxetine versus placebo in the treatment of pain: BPI severity ‐ average pain.

Heterogeneity

Inclusion of data contributed by Gao 2010 caused heterogeneity in meta‐analyses. Where this was the case we repeated the analyses excluding the Gao 2010 data. The heterogeneity was probably the result of the estimated final doses of duloxetine used in the analysis figures.

Significant heterogeneity was not otherwise present except for the SF‐36 physical component summary, PGIC, bodily pain index and pain at rest. Heterogeneity was present in the subgroup analyses and also in the 'all doses' analysis, where doses were combined. The origin of this heterogeneity was not always clear and therefore we performed these analyses with a random‐effects model.

Sensitivity analysis

We attempted prespecified sensitivity analyses. All trials were carried out by the drug manufacturer and all had more than 100 participants. However, only three of the included studies had a dropout rate of less than 20%. When we included only these studies in the analysis, although duloxetine remained significantly effective (number of participants achieving ≥ 50% reduction in pain RR 1.83 (95% CI 1.41 to 2.36), the effect of duloxetine at 120 mg was lost and the effect of duloxetine at all doses was barely significant (RR 1.55, 95% CI 1.01 to 2.38).

Painful peripheral neuropathy ‐ duloxetine versus pregabalin

In the only comparison of duloxetine and pregabalin (Tesfaye 2013) (804 participants), the proportion of participants responding to duloxetine 60 mg by achieving 50% or more reduction in pain was significantly greater than those responding to pregabalin 300 mg daily (RR 1.46, 95% CI 1.19 to 1.80) (see Analysis 2.1). Both doses represent realistic therapeutic target doses for treatment. The magnitude of change was also greater for duloxetine than pregabalin (RR ‐0.62, 95% CI ‐0.92 to ‐0.32) (see Analysis 2.2). The number improved by 30% or more at 12 weeks was significantly greater with duloxetine than placebo (RR 1.42, 95% CI 1.20 to 1.68) (see Analysis 2.3).

The response rate for a ≥ 50% reduction in pain for duloxetine was 38%, whereas the 26% response rate to pregabalin was approximately the same as the placebo response rate in the other trials of duloxetine at 60 mg (compare Analysis 1.1 and Analysis 2.1). This raises questions about the similarity of the selected groups of participants or the efficacy of pregabalin, which is known, however, to be effective in other studies.

Painful peripheral neuropathy ‐ duloxetine versus amitriptyline

The only trial comparing duloxetine to amitriptyline (Kaur 2011) was a blinded cross‐over study with 62 participants comparing six weeks treatment with each active agent in escalating dose: duloxetine up to 60 mg and amitriptyline up to 50 mg. The trial did not meet our predefined inclusion criteria of eight weeks of study medication but is included here for completeness as it is the only comparative trial of its type. Significant carryover effects were evident (VAS pain scores only returned to 75% of baseline during washout (see Figure 2 of the Kaur 2011 paper)). In addition, a number of predefined outcome measures were not presented in the results, so there was an unclear risk of bias. Sixty‐five per cent of participants achieved 60 mg of duloxetine per day and 48% of participants 50 mg amitriptyline. The majority of participants (59% duloxetine and 55% amitriptyline) were reported to have achieved a 'good' (> 50% improvement) response to the interventions.

It is not possible to re‐analyse the data as no raw data are available. We contacted the authors by email to provide original data to enter into a GIV analysis. No reply was forthcoming.

Trial sequential analysis in painful peripheral neuropathy

We performed TSA for the primary outcome of a ≥ 50% reduction in pain at 12 weeks or less with at least 8 weeks of treatment with duloxetine, for the trials that compared duloxetine to placebo. There was not enough information to explore the pairwise trials. The TSA report demonstrated that although the optimal information size had not been reached, the Z‐score favoured duloxetine and diverged from futility (Figure 7).


Trial sequential analysis of duloxetine versus placebo in the treatment of painful neuropathy ‐ 50% or more reduction in pain at 8‐12 weeks with at least 8 weeks of treatment

Trial sequential analysis of duloxetine versus placebo in the treatment of painful neuropathy ‐ 50% or more reduction in pain at 8‐12 weeks with at least 8 weeks of treatment

Fibromyalgia ‐ duloxetine versus placebo

Primary outcome

FIve trials reported data corresponding to the primary outcome for this review (Arnold 2004; Arnold 2005; Russell 2008; Arnold 2010; Arnold 2012) and the sixth reported data for the same outcome at more than 12 weeks (Chappell 2008). The studies used two scales; in Arnold 2004, the Fibromyalgia Impact Questionnaire pain score (Burckhardt 1991), and in the remainder, the BPI modified SF‐36 average pain severity score. The 20 mg dose of duloxetine in Russell 2008 (223 participants) did not show significant differences in any of the reported measures. The 30 mg dose, used only in Arnold 2012 (308 participants), was also negative on all measures of outcome except for a statistically significant benefit on the Patient Global Impression of Improvement (PGI‐I) (interchangeable with the Patient Global Impression of Change (PGI‐C)) and the mental component ot the SF‐36, neither of which were of a magnitude to be clinically significant. Both of these studies had wide CIs because of the small number of participants (Analysis 3.1). Five studies reported short‐term (up to and including 12 weeks) ≥ 50% improvement of pain compared with baseline. The RR of improvement was significantly greater with duloxetine 60 mg (1.57, 95% CI 1.20 to 2.06; 2 trials, 528 participants) and with 120 mg daily (1.69, 95% CI 1.40 to 2.03; 4 trials, 1234 participants) than with placebo (see Analysis 3.1). The RR of improvement compared with placebo for all doses in all five short‐term trials, which had a total of 1887 participants, was 1.50 (95% CI 1.29 to 1.75). Exclusion of the data for 111 participants in the Arnold 2012 study, which was responsible for the heterogeneity in the analysis, gave an RR of 1.68 (95% CI 1.41 to 2.01).

Secondary outcomes

Two studies looked at long‐term outcomes at more than 12 weeks (Chappell 2008; Russell 2008). These investigators documented outcomes at 28 weeks. Improvement of pain ≥ 50% compared with baseline at 27 to 28 weeks was similar between the 60 mg and 120 mg doses (no dose effect) and when all 989 participants were combined, despite the Chappell trial being negative, the RR for improvement was 1.40 (95% CI 1.09 to 1.79) (see Analysis 3.2).

The RR of ≥ 30% improvement at 12 weeks or less was significantly greater than placebo with the duloxetine 60 mg dose (RR 1.52, 95% CI 1.24 to 1.85; 2 trials, 528 participants) and 120 mg dose (RR 1.46, 95% CI 1.26 to 1.69; 3 trials, 1020 participants) but not with the 20 mg or 30 mg doses (see Figure 8, Analysis 3.3). The RR for all doses combined was 1.38 (95% CI 1.22 to 1.56; 4 trials, 1673 participants). It is notable that no dose effect exists from 60 mg to 120 mg. There was no statistically significant improvement in pain at 30 mg duloxetine in the only trial presenting data for the mean improvement but at 120 mg there was a significant benefit in favour of duloxetine (MD ‐0.80, 95% CI ‐1.35 to ‐0.25; 1 trial, 507 participants) (see Analysis 3.4).


Duloxetine versus placebo in the treatment of fibromyalgia: >30% improvement <12 weeks.

Duloxetine versus placebo in the treatment of fibromyalgia: >30% improvement <12 weeks.

All six studies documented the physical component summary scores and bodily pain subscores of the SF‐36 and most reported the bodily pain subscore (Arnold 2004; Arnold 2005; Chappell 2008; Russell 2008; Arnold 2010; Arnold 2012). For the mental component summary score, the 30 mg, 60 mg and 120 mg doses had increasing effect compared to placebo (for the 120 mg dose, MD 4.22, 95% CI 2.43 to 6.02; 5 trials, 1531 participants) (see Analysis 3.5). Interestingly, the physical component summary score was only significant at the 120 mg dose of duloxetine (MD 2.13, 95% CI 0.95 to 3.30; 5 trials, 1531 participants) (see Analysis 3.6). For the bodily pain subscale, the RR of improvement from four studies (Arnold 2004; Arnold 2005; Chappell 2008; Arnold 2010) was significantly greater for duloxetine than placebo at both the 60 mg dose (MD 8.20, 95% CI 3.20 to 13.20; 1 trial, 221 participants) and the 120 mg dose (MD 5.96, 95% CI 3.76 to 8.16; 4 trials, 1243 participants) (Figure 9, Analysis 3.7). Again, it is notable that the 120 mg dose had less effect than the 60 mg dose.


Duloxetine versus placebo in the treatment of fibromyalgia: SF‐36 bodily pain.

Duloxetine versus placebo in the treatment of fibromyalgia: SF‐36 bodily pain.

Four studies reported the PGI‐I (Arnold 2005; Russell 2008; Chappell 2008; Arnold 2012), which was significantly in favour of duloxetine at the 20 mg, 30 mg, 60 mg and 120 mg doses (120 mg dose MD ‐0.44, 95% CI ‐0.66 to ‐0.23; 3 trials, 826 participants) (see Analysis 3.8). The magnitude of change at each dose was very similar, with no dose effect. However, the magnitude of change failed to reach a level considered to be clinically significant.

Sensitivity analysis

No data were suitable for the prespecified sensitivity analyses.

Trial sequential analysis in fibromyalgia

We performed TSA on the primary outcome data for ≥ 50% reduction in pain at ≤ 12 weeks with at least eight weeks of treatment with 60 mg duloxetine (the standard dose). WIth the data so far available, the Z‐score only just crossed the boundary of significance, although it was divergent from futility (Figure 10). The optimal information size is some way off and more trials and participants are required to make convincing statements about the efficacy of duloxetine for this indication at this dose.


Trial sequential analysis of duloxetine 60 mg versus placebo for the 50% reduction in pain in fibromyalgia with at least 8 weeks treatment at 8‐12 weeks

Trial sequential analysis of duloxetine 60 mg versus placebo for the 50% reduction in pain in fibromyalgia with at least 8 weeks treatment at 8‐12 weeks

Painful physical symptoms not explained by any known alternative diagnosis in the context of major depressive disorder ‐ duloxetine versus placebo

Primary outcome

Painful physical symptoms associated with major depressive disorder have been assessed in three studies each lasting eight weeks that used a duloxetine dose of 60 mg daily (Brecht 2007; Gaynor 2011a; Gaynor 2011b). The proportion of participants achieving ≥ 50% pain relief was greater with duloxetine than placebo in two studies (1023 participants) for which adequate information was available (RR 1.37, 95% CI 1.19 to 1.59) (see Analysis 4.1) and the magnitude of improvement was greater in those two studies (MD ‐0.55, 95% CI ‐0.75 to ‐0.35) (see Analysis 4.3). No data were available for Brecht 2007 in the comparison of magnitude as the report does not provide SDs, although the absolute magnitude of improvement was similar to the two studies of Gaynor.

Secondary outcomes

More participants improved ≥ 30% in their levels of pain than with placebo at 60 mg duloxetine per day (RR 1.27, 95% CI 1.15 to 1.40; 3 studies, 1359 participants (see Analysis 4.2).

No data were available for the subscores of the SF‐36 or to calculate a mean improvement in the PGI‐I scores.

Sensitivity analysis

No data were suitable for the prespecified sensitivity analyses.

Trial sequential analysis in painful physical symptoms in depression

TSA was performed on the primary outcome data for ≥ 50% reduction in pain at <12 weeks with at least 8 weeks of treatment with 60 mg duloxetine (the standard dose). The optimal information size was exceeded but there were not enough data to calculate an area of futility (Figure 11). However, there is convincing evidence from the small number of studies that duloxetine is efficacious.


Trial Sequential Analysis of duloxetine 60 mg versus placebo in the treatment of painful physical symptoms in depression at less than 12 weeks with at least eight weeks of treatment

Trial Sequential Analysis of duloxetine 60 mg versus placebo in the treatment of painful physical symptoms in depression at less than 12 weeks with at least eight weeks of treatment

Central neuropathic pain ‐ duloxetine versus placebo

A single investigator‐led study with a low risk of bias but only 48 participants looked at the effect of duloxetine in people with central neuropathic pain (Vranken 2011). This was the only trial not sponsored or run by the company manufacturing duloxetine. However, it was the smallest of the studies and only one of three with no significant risk of bias.

There was no therapeutic effect of duloxetine on the neuropathic pain of the participants in this study on any of our pre‐defined outcome measures that the study reported (see Analysis 5.1; Analysis 5.2; Analysis 5.3; Analysis 5.4). There was a borderline effect in the bodily pain domain of the SF‐36 (MD 8.00, 95% CI ‐0.81 to 16.81), which did not reach significance, and the proportion of participants reporting improvement on the PGI‐I was just significant (RR 2.75, 95% CI 1.02 to 7.44) (see Analysis 5.4 and Analysis 5.5). The trial also reported a statistically significant improvement in the severity of dynamic and cold allodynia. Given that the trial was small, the trial authors recommended that more studies of central neuropathic pain are performed.

Sensitivity analysis

Vranken 2011 was the only trial of central neuropathic pain and no meta‐analysis or sensitivity analysis was therefore possible.

Trial sequential analysis ‐ central neuropathic pain

We did not perform TSA on the single study available.

Adverse events (all indications)

We analysed adverse events across all included studies (all indications).

Serious adverse events were uncommon and were no more frequent with duloxetine than placebo at any dose or when combining all doses together (42 events in 2785 duloxetine‐treated participants versus 39 events in 2191 placebo participants RR 0.81 (95% CI 0.53 to 1.25) (see Analysis 6.7).

Adverse events of any sort, however, were very common in all of the trials in both experimental and placebo groups.

The rate of any adverse event was high in both the treatment and placebo arms of all studies, with 1530 adverse events being reported in 2462 control participants and 2033 adverse events occurring in 2796 participants in the combined treatment arms covering all doses (RR 1.15, 95% CI 1.11 to 1.20) (see Analysis 6.1). Adverse events were significantly more common with duloxetine than with placebo especially in 60 mg (RR 1.15, 95% CI 1.10 to 1.20) and 120 mg (RR 1.19, 95% CI 1.09 to 1.30) duloxetine groups (Analysis 6.1). Doses of 60 mg and 120 mg duloxetine were also associated with a significantly greater risk of cessation compared to placebo (Analysis 6.6, Figure 12).


Adverse events leading to cessation of treatment.

Adverse events leading to cessation of treatment.

The most common individual adverse events were nausea (Analysis 6.2), dry mouth (Analysis 6.3), dizziness (Analysis 6.4), somnolence (Analysis 6.5), fatigue, insomnia, constipation, decreased appetite, sweating and rhinitis. All had a dose dependency, with a greater frequency of side effects at 120 mg daily than 60 mg daily. No suicides were reported where suicide and risk were mentioned; this outcome was rarely specifically sought.

Discussion

This updated Cochrane Systematic Review of duloxetine for the treatment of chronic pain and fibromyalgia identified 12 more studies than the original review that fitted the predefined quality criteria for inclusion, bringing the number of included studies to 18. These studies covered painful peripheral neuropathy (diabetic), central neuropathic pain, fibromyalgia, and painful physical symptoms in people with major depressive disorder and no underlying explanation for their pain. We excluded 25 studies identified in our searches for various reasons; many concerned pain from other conditions (for example, pelvic pain or osteoarthritis of the knee) that are the topics of other reviews. Some did not fulfil our criteria of treating participants for at least eight weeks (e.g. Brannan 2005), some were open label (e.g. Raskin 2006a and Raskin 2006b) and some not randomised or controlled (Canovas 2007). One trial comparing a novel agent ADL‐5859 to duloxetine and placebo has been completed but remained unpublished at the time of writing; no data were available from the company. Five studies are ongoing (NCT00457730; NCT00619983; NCT01179672; NCT01237587; NCT01552057) and the results of these are likely to become available in due course. Following extensive searches of ClinicalTrials.gov and reference databases, handsearching reference lists, cross‐correlating NCT codes, titles and abstracts, the authors did not identify any further trials. The Lilly Trials Database is freely available and contains extensive details of all trials. Unfortunately, neither the ClinicalTrials.gov NCT number nor the final published title of the research are published in the Lilly database, which make it very difficult to check whether all trials registered on ClinicalTrials.gov have been published. We have no reason to suspect extensive publication bias, certainly in the last few years, but we could not find publications corresponding to five ClinicalTrials.gov entries (Appendix 6).

We analysed the effects of duloxetine on painful diabetic neuropathy, central pain, fibromyalgia, and painful physical symptoms in depression separately. Each has a different pathogenesis and hence analysis of all conditions together would be meaningless. Furthermore, patients and caregivers are more likely to glean benefit from data presented for individual diseases. There is then the potential, should the need arise, to extrapolate such data as a guide to similar diseases where no evidence exists. In meta‐analyses, the magnitude of the benefit in terms of pain relief was similar in all the individual conditions.

For painful diabetic neuropathy, the RR of ≥ 50% reduction in pain at eight to 12 weeks at all doses of duloxetine versus placebo was 1.53 (95% CI 1.21 to 1.92) (NNTB 7 (5 to 10)). For the standard dosage of 60 mg duloxetine daily, this corresponds to an NNTB of 5 (95% CI 4 to 7) (summary of findings Table for the main comparison). For ≥ 30% improvement of pain, the RR at 60 mg was 1.53 (95% CI 1.33 to 1.75), corresponding to an NNTB of 5 (95% CI 4 to 8) (summary of findings Table for the main comparison). These NNTBs are of similar magnitude to the NNTB for tricyclic antidepressants for the achievement of moderate pain relief (NNTB 3.6, 95% CI 3 to 4.5) (Saarto 2007; Kajdasz 2007), and for amitriptyline for 'improvement' (NNTB) of 4.6 (95% CI 3.6 to 6.6) (Moore 2012). The mean difference in pain at a dose of 60 mg duloxetine daily was ‐0.96 points (95% CI ‐1.26 to ‐0.65 points) compared with placebo, on an 11‐point scale. A small dose‐effect may be present, but in most outcome measures, 20 mg to 30 mg is not clearly effective, and 40 mg to 120 mg is effective, with a very small efficacy increment as the dose is increased. There were improvements in the other prespecified secondary outcome measures at 40 mg, 60 mg, and 120 mg daily doses of duloxetine, again with slightly but not significantly greater improvements with the 120 mg dose. The PGIC secondary outcomes were statistically significant, but only border on magnitudes of change that are currently considered to be clinically significant (Dworkin 2008). We discovered no RCTs of the effect of duloxetine on painful diabetic neuropathy for periods longer than 12 weeks.

A number of studies exist presenting the minimal clinically important difference (MCID) for pain in various painful conditions on a 100 mm VAS, which can be roughly translated into an 11‐point VAS. Studies vary in design and inclusion criteria, and the MCID or minimal clinically important change varies with the position on the scale from which participants start. However, for people with pain measured in the middle of the VAS (moderate pain) at baseline, MCID is in the region of two to three points on an 11‐point scale (Mease 2011; Salaffi 2004). Changes of one point or less are unlikely to be clinically relevant. However, as with all pain studies, it is recognised that there is a U‐shaped response curve, with some people responding maximally and some not at all. This makes globalised linear measures of improvement moderately meaningless as the lack of response in some dilutes the effect that occurs in others. The proportion of people responding becomes a more useful measure.

For participants with fibromyalgia, the magnitude of improvement in pain at 12 weeks was similar to that seen in participants with diabetic peripheral neuropathy. There was no clear difference between the effects of duloxetine in fibromyalgia or diabetic peripheral neuropathy when assessed on the primary outcome measures, and so even if fibromyalgia is a "different sort of pain" (Dadabhoy 2006), it seems to respond to duloxetine in a similar way. However, the absolute risk reduction was marginally less than that for diabetic peripheral neuropathic pain and the corresponding NNTB for ≥ 50% or more pain relief at a duloxetine dose of 60 mg daily was 8 (95% CI 5 to 17) (summary of findings Table 2). When we combined results for all doses, the NNTB was 9 (95% CI 7 to 13). Although there were fewer data for fibromyalgia, there again seemed to be a floor effect of dosage, where 30 mg was not effective but higher doses appeared to be. The ceiling effect (or lack of any additional therapeutic effect at 120 mg) was not evident as there were too few data.

It is notable that in fibromyalgia the magnitude of improvement in the SF‐36 mental subscore (MD, 120 mg dose, 4.22, 95% CI 2.43 to 6.02) was double that of peripheral neuropathic pain at the same dose (MD 2.23, 95% CI 0.69 to 3.77) or central neuropathic pain; whereas the magnitude of improvement in bodily pain scores in the neuropathic pain studies (MD 8.19, 95% CI 4.33 to 12.05) was 40% more than that in fibromyalgia (MD 5.96, 95% CI 3.76 to 8.16). As is noted above, the authors of Brecht 2007 comment on the difference in tempo of improvement of depression and pain scores, suggesting that different, but similar, mechanisms are responsible for the two phenomena. In Gaynor 2011a "..patients who met the >30% or >50% BPI response criteria at the 8 week LOCF endpoint had rates of [depression] remission (MADRS total scores…) that were higher compared to duloxetine treated patients who did not meet the BPI response criterion", and in Gaynor 2011b "..remission rates [of depression] were three times greater for patients taking duloxetine who reported at least 30% reduction in pain versus those who did not." However, in Raskin 2005, depression scales did not change despite similar pain scale improvements but the trial specifically excluded people with pre‐existing depression from entry. Russell 2008 used two regression models to separate the direct effect of duloxetine on pain and the indirect effect on the treatment of depression in the treatment of pain. The trial authors calculated that between 21% and 38% of the pain treatment effect was due to treatment of depressive symptoms. Fava 2004 estimated a 50% influence. This remains circumstantial evidence that the mechanisms of pain relief involve both mood and direct pain components and where mood is more involved there may be a greater influence on pain relief.

This new version of the review contains two large, new studies including people with major depressive symptoms and pain of unclear origin. Although these studies were generally well performed, limitations of allocation concealment, blinding and randomisation, along with the potential heterogeneous mix of causes of pain (including various pains of unknown origin), potentially leads to extensive imprecision and a lack of certainty about the results achieved. The mean improvement in pain was only ‐0.55 points on an 11‐point VAS (95% CI ‐0.75 to ‐0.35). The RR of people achieving 50% or more relief of pain at under 12 weeks was 1.37 (95% CI 1.19 to 1.59; NNTB 8, 95% CI 5 to 14) (summary of findings Table 3), once again of a similar magnitude to the pain in fibromyalgia. Unfortunately, there were no data on the SF‐36 to make further comparisons.

This updated review also contains a single study independent from the makers of duloxetine exploring the effect of duloxetine versus placebo in the treatment of central neuropathic pain from spinal cord disorders or stroke. This single, high quality, independent and well‐performed study suffers from its small size and so effect estimates are associated with large CIs. The only result of statistical significance was the PGIC, with a RR favouring duloxetine that was just statistically significant at 2.75 (95% CI 1.02 to 7.44). The magnitude of improvement in pain was also similar to other conditions, although it was nonsignificant, with wide CIs.

A number of studies were not included in the meta‐analysis and are included here for completeness. Kaur 2011 performed a randomised cross‐over trial of amitriptyline and duloxetine. It was not included in a formal meta‐analysis because it was the only trial comparing those interventions. It did not meet our predefined inclusion criteria for length but we included it because it was the only one of its type. It had significant carryover between the cross‐over arms, which greatly affected quality. Duloxetine has a superior response to pregabalin in Tesfaye 2013, but the pregabalin response was at the level of a placebo response in other trials; other studies (for example, Moore 2009) suggest that pregabalin is effective and hence the comparative benefit of duloxetine over placebo remains unclear. Brannan 2005 performed a double‐blind RCT of duloxetine in major depressive disorders, in which they also measured the effect on pain. The outcomes were measured at seven weeks and hence it did not meet our eligibility criteria. Furthermore, the placebo and active arms of the trial were not balanced for depression as measured by the Hamilton Rating Scale for Depression at entry. However, depression scores did not change significantly and similar improvements were seen in pain scores as in the fibromyalgia and painful diabetic neuropathy trials. This compares favourably with the longer studies of Gaynor (Gaynor 2011a; Gaynor 2011b). The Raskin 2006a trial was a 52‐week extension phase of the prior 13‐week study (Raskin 2005) but although this trial was randomised, it was open, with a non‐standardised control group ("standard care"). Hence we excluded it. The only subscale outcomes of the SF‐36 eligible for this review from this trial did not change over one year, but duloxetine was safe and well‐tolerated over that period. Wernicke 2006b was a similar randomised but open extension phase study of Goldstein 2005. Again, safety and tolerability were the main focus. The SF‐36 bodily pain subscore improved significantly but by a much smaller magnitude than in the studies included in the meta‐analysis. Finally, the Goldstein 2004 study reported, with a dose of duloxetine of 80 mg, a median improvement in pain severity on a VAS for pain of ‐7.5% (interquartile range ‐25% to 1%), assessed at week eight. This was the only pain measure to show statistically significant improvement. Since pain was not an inclusion criterion for the trial (designed primarily to look at depression), the trial did not match our pre‐specified inclusion criteria. Hence it was not included in the analysis above but adds support to the therapeutic efficacy of duloxetine in the treatment of pain.

We included TSA in this review for the first time. These analyses are useful in a number of ways. There was not enough information to create a TSA for central pain. For the primary outcome in painful diabetic neuropathy, treatment with the standard dose of duloxetine had convincing evidence of non‐futility even though the optimal information size had not been reached. Within the predefined parameters, even after the first two studies the Z‐score had crossed the boundary line of efficacy. It remains that none of these trials was performed by an independent investigator, but the trials were generally of high to moderate quality and there was no reason to suspect that further independent trials would significantly skew the result to futility. In fibromyalgia, the optimal information size was some way off and further independent trials are recommended. For painful physical symptoms associated with depression, the optimal information size had been exceeded but there were insufficient data to calculate an area of futility. However, the efficacy in this indication is clear. As indicated above, the magnitude of improvement in terms of MD should be considered.

Adverse events were very common in these trials but were, in general, mild. The rates of any adverse event and adverse events leading to cessation of treatment were significantly greater with duloxetine than with placebo at the 60 mg and 120 mg doses, which are the doses used in clinical practice. However, withdrawals because of adverse events were relatively few (duloxetine, all doses combined, 12.6% versus placebo 5.8% (RR 1.99, 95% CI 1.67 to 2.37)). The NNTH for cessation of treatment was 17 (95% CI 13 to 26). These figures are in line with the retrospective analysis of Gahimer et al. in 23,983 patients in the duloxetine integrated exposures database (Gahimer 2007), where approximately 20% of patients withdrew because of adverse events. Adverse event rates were also dose related, being more common with the 120 mg than the 60 mg dose and with the 60 mg than the 20 mg or 30 mg doses, whether any adverse event or events leading to cessation are considered. The rates of serious adverse events were not greater with duloxetine than with placebo (RR 0.81, 95% CI 0.53 to 1.25). The adverse event profile was broadly in line with adverse events in the Cochrane Systematic Review of duloxetine in stress incontinence (Mariappan 2009). From observational studies, the most common side effects of duloxetine are quoted as nausea (37%), dry mouth (32%), dizziness (22%), somnolence (20%), insomnia (20%) and diarrhoea (14%) (Aronson 2007). In the studies included here, sweating (60 mg daily 6.5%, 120 mg daily 9.3%, versus control 0.88%) was also common, occurring at about the same frequency as reported by Gahimer et al. (6.2%) (Gahimer 2007). Tremor was also commonly reported, especially at the 120 mg dose in one trial (60 mg daily 3.3%, 120 mg daily 10.2%) (Russell 2008). There were small changes in blood pressure and heart rate in some but not all of the included studies, although we have not formally analysed these. However, in a review of the trials in depression, duloxetine was reported to produce a small, statistically significant rise in heart rate (2 beats per minute) and a sustained increase in systolic blood pressure versus placebo (1% rise versus 0.4%) (Aronson 2008). A company‐performed retrospective database review of 8504 participants in 42 placebo‐controlled studies of duloxetine covering five indications identified no significant cardiovascular risk (Wernicke 2007). There were no significant increases in suicide or suicidal thoughts in studies where these event were reported.

The studies included in this meta‐analysis contribute more than 6400 participants to meta‐analyses of the use of duloxetine in neuropathic pain, fibromyalgia and painful symptoms in depressive illness, and this is the most comprehensive assessment of any individual antidepressant drug for these indications of which we are aware. This provides a greater level of certainty to the conclusions of this review. Other tricyclic antidepressants have greater efficacy in terms of lower NNTBs than duloxetine, but the trials are small, many have methodological deficiencies and so efficacy may be overestimated. The European Federation of Neurological Societies (EFNS) guidelines for the pharmacological treatment of neuropathic pain recommend duloxetine as second line to tricyclic antidepressants and gabapentin or pregabalin, except where cardiovascular risk factors are present, when duloxetine is favoured (Attal 2006). The National Institue for Health and Clinical Excellence (NICE) recommends duloxetine as first line therapy in diabetic neuropathic pain in non‐specialist care settings but fails to mention it in the treatment of other painful neuropathies (http://publications.nice.org.uk/neuropathic‐pain‐cg96/guidance).

The cost effectiveness of duloxetine has not been formally investigated in RCTs. Four studies have addressed cost effectiveness (Wu 2006; Beard 2008; O'Connor 2008, Bellows 2012); the company manufacturing the drug performed two of them (Wu 2006; Beard 2008). The quality of the earlier studies was questionable but the latest independent study estimates similar levels of economic cost associated with duloxetine use. Wu 2006 performed an analysis of the cost effectiveness of duloxetine in participants completing the Goldstein 2005 trial in the USA. Using trial outcomes, the study authors compared the costs to healthcare, society and employers with those of the 'standard therapies' in the control group. Given that duloxetine was shown to be more effective than standard therapies, duloxetine was also shown to significantly reduce societal and employer costs with a trend towards cost effectiveness in medical costs when compared to standard treatment with other pain management therapies. Beard 2008 used a decision analytic model to represent the sequential management of people with diabetic peripheral neuropathic pain based upon current prescribing practice in the UK. Calculations in the model were based upon data for clinical efficacy that are good for duloxetine and pregabalin, but less robust for tricyclic antidepressants and gabapentin ('first line' drugs). However, recognising the inherent limitations, the authors calculated that duloxetine added as a second line therapy resulted in a predicted cost saving of GBP 77 per patient, on the basis that an additional 29 patients per 1000 achieved a full pain response compared to standard treatment. When duloxetine is added to the standard prescribing hierarchy as a second line therapy, the clinical benefit can also be expressed as adding an additional 0.0019 quality‐adjusted life years (QALYs) per patient. O'Connor 2008 also used a decision analytic model incorporating published and unpublished data from RCTs and cross‐sectional studies of duloxetine, gabapentin, pregabalin and desipramine. The QALY cost of duloxetine using this model varied from USD 47,700 to USD 867,000 per QALY depending upon the assumptions made in the analysis of the trial data. In the latest study, Bellows 2012 used a decision tree model in both clinical trial and real world studies comparing duloxetine to pregabalin. Duloxetine demonstrated an incremental cost of minus USD 187 per patient or incremental effectiveness of 0.011 QALYs. At a cost per QALY threshold of 50,000 USD (almost equivalent to the UK's GBP 30,000), duloxetine is more cost effective than pregabalin. This latter analysis presents the best case scenario, since although the cost effectiveness in people with good pain relief is high, the cost effectiveness can be overwhelmed by those patients in whom a drug does not work, and so cost effectiveness studies of this sort are questionable.

Methodological quality summary: review authors' judgements about each methodological quality item for each included study. Green = low risk of bias; yellow = unclear risk of bias; red = high risk of bias
Figuras y tablas -
Figure 1

Methodological quality summary: review authors' judgements about each methodological quality item for each included study. Green = low risk of bias; yellow = unclear risk of bias; red = high risk of bias

Duloxetine versus placebo in the treatment of painful neuropathy: Number of patients with >50% improvement of pain at <12 weeks.
Figuras y tablas -
Figure 2

Duloxetine versus placebo in the treatment of painful neuropathy: Number of patients with >50% improvement of pain at <12 weeks.

Duloxetine versus placebo in the treatment of pain: Mean improvement in pain at 12 weeks.
Figuras y tablas -
Figure 3

Duloxetine versus placebo in the treatment of pain: Mean improvement in pain at 12 weeks.

Duloxetine versus placebo in the treatment of pain: Number of patients with >30% improvement in pain at <12 weeks.
Figuras y tablas -
Figure 4

Duloxetine versus placebo in the treatment of pain: Number of patients with >30% improvement in pain at <12 weeks.

Duloxetine versus placebo in the treatment of pain: Patient reported global impression of change.
Figuras y tablas -
Figure 5

Duloxetine versus placebo in the treatment of pain: Patient reported global impression of change.

Duloxetine versus placebo in the treatment of pain: BPI severity ‐ average pain.
Figuras y tablas -
Figure 6

Duloxetine versus placebo in the treatment of pain: BPI severity ‐ average pain.

Trial sequential analysis of duloxetine versus placebo in the treatment of painful neuropathy ‐ 50% or more reduction in pain at 8‐12 weeks with at least 8 weeks of treatment
Figuras y tablas -
Figure 7

Trial sequential analysis of duloxetine versus placebo in the treatment of painful neuropathy ‐ 50% or more reduction in pain at 8‐12 weeks with at least 8 weeks of treatment

Duloxetine versus placebo in the treatment of fibromyalgia: >30% improvement <12 weeks.
Figuras y tablas -
Figure 8

Duloxetine versus placebo in the treatment of fibromyalgia: >30% improvement <12 weeks.

Duloxetine versus placebo in the treatment of fibromyalgia: SF‐36 bodily pain.
Figuras y tablas -
Figure 9

Duloxetine versus placebo in the treatment of fibromyalgia: SF‐36 bodily pain.

Trial sequential analysis of duloxetine 60 mg versus placebo for the 50% reduction in pain in fibromyalgia with at least 8 weeks treatment at 8‐12 weeks
Figuras y tablas -
Figure 10

Trial sequential analysis of duloxetine 60 mg versus placebo for the 50% reduction in pain in fibromyalgia with at least 8 weeks treatment at 8‐12 weeks

Trial Sequential Analysis of duloxetine 60 mg versus placebo in the treatment of painful physical symptoms in depression at less than 12 weeks with at least eight weeks of treatment
Figuras y tablas -
Figure 11

Trial Sequential Analysis of duloxetine 60 mg versus placebo in the treatment of painful physical symptoms in depression at less than 12 weeks with at least eight weeks of treatment

Adverse events leading to cessation of treatment.
Figuras y tablas -
Figure 12

Adverse events leading to cessation of treatment.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.
Figuras y tablas -
Analysis 1.1

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 1.2

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 3 Number of participants with ≥ 30% improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 1.3

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 3 Number of participants with ≥ 30% improvement in pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 4 Mean improvement in SF‐36 Physical Subscore at 12 weeks or less.
Figuras y tablas -
Analysis 1.4

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 4 Mean improvement in SF‐36 Physical Subscore at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 5 Mean improvement in SF‐36 Mental Subscore at 12 weeks or less.
Figuras y tablas -
Analysis 1.5

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 5 Mean improvement in SF‐36 Mental Subscore at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 6 Mean improvement in SF‐36 Bodily Pain Subscore at 12 weeks or less.
Figuras y tablas -
Analysis 1.6

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 6 Mean improvement in SF‐36 Bodily Pain Subscore at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 7 Mean improvement in Patient Reported Global Impression of Improvement at 12 weeks or less.
Figuras y tablas -
Analysis 1.7

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 7 Mean improvement in Patient Reported Global Impression of Improvement at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 8 Mean improvement in BPI Severity ‐ average pain at 12 weeks or less.
Figuras y tablas -
Analysis 1.8

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 8 Mean improvement in BPI Severity ‐ average pain at 12 weeks or less.

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 9 Mean improvement in pain at rest (night pain) at 12 weeks or less.
Figuras y tablas -
Analysis 1.9

Comparison 1 Duloxetine versus placebo in the treatment of painful diabetic neuropathy, Outcome 9 Mean improvement in pain at rest (night pain) at 12 weeks or less.

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 2.1

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 1 Number of participants with ≥ 50% improvement in pain at 12 weeks or less.

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 2.2

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 2 Mean improvement in pain at 12 weeks or less.

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 3 Number improved ≥ 30% at 12 weeks or less.
Figuras y tablas -
Analysis 2.3

Comparison 2 Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy, Outcome 3 Number improved ≥ 30% at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.
Figuras y tablas -
Analysis 3.1

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 2 Number of participants with ≥ 50% improvement of pain at more than 12 weeks.
Figuras y tablas -
Analysis 3.2

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 2 Number of participants with ≥ 50% improvement of pain at more than 12 weeks.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 3 Number of participants with ≥ 30% improvement of pain at 12 weeks or less.
Figuras y tablas -
Analysis 3.3

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 3 Number of participants with ≥ 30% improvement of pain at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 4 Mean improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 3.4

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 4 Mean improvement in pain at 12 weeks or less.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 5 Mean improvement in the SF‐36 mental component summary subscore.
Figuras y tablas -
Analysis 3.5

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 5 Mean improvement in the SF‐36 mental component summary subscore.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 6 Mean improvement in the SF‐36 physical component summary subscore.
Figuras y tablas -
Analysis 3.6

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 6 Mean improvement in the SF‐36 physical component summary subscore.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 7 Mean improvement in the SF‐36 Bodily Pain Subscore.
Figuras y tablas -
Analysis 3.7

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 7 Mean improvement in the SF‐36 Bodily Pain Subscore.

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 8 Mean improvement in the Patient reported Global Impression of Change at completion of trial.
Figuras y tablas -
Analysis 3.8

Comparison 3 Duloxetine versus placebo in the treatment of fibromyalgia, Outcome 8 Mean improvement in the Patient reported Global Impression of Change at completion of trial.

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 1 Number of participants with > 50% pain relief at 12 weeks or less.
Figuras y tablas -
Analysis 4.1

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 1 Number of participants with > 50% pain relief at 12 weeks or less.

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 2 Participants with > 30% pain relief at 12 weeks or less.
Figuras y tablas -
Analysis 4.2

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 2 Participants with > 30% pain relief at 12 weeks or less.

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 3 Mean improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 4.3

Comparison 4 Duloxetine versus placebo for the treatment of pain in major depressive disorder, Outcome 3 Mean improvement in pain at 12 weeks or less.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 1 Mean improvement in pain at 12 weeks or less.
Figuras y tablas -
Analysis 5.1

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 1 Mean improvement in pain at 12 weeks or less.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 2 Mean improvement in SF‐36 Physical Subscore.
Figuras y tablas -
Analysis 5.2

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 2 Mean improvement in SF‐36 Physical Subscore.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 3 Mean improvement in the SF‐36 Mental Subscore at 12 weeks.
Figuras y tablas -
Analysis 5.3

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 3 Mean improvement in the SF‐36 Mental Subscore at 12 weeks.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 4 Mean improvement in the SF‐36 Bodily Pain Subscore.
Figuras y tablas -
Analysis 5.4

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 4 Mean improvement in the SF‐36 Bodily Pain Subscore.

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 5 Number of participants improved on PGI‐I (better or very much better).
Figuras y tablas -
Analysis 5.5

Comparison 5 Duloxetine versus placebo in the treatment of central neuropathic pain, Outcome 5 Number of participants improved on PGI‐I (better or very much better).

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 1 Proportion of participants with any adverse event.
Figuras y tablas -
Analysis 6.1

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 1 Proportion of participants with any adverse event.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 2 Nausea.
Figuras y tablas -
Analysis 6.2

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 2 Nausea.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 3 Dry mouth.
Figuras y tablas -
Analysis 6.3

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 3 Dry mouth.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 4 Dizziness.
Figuras y tablas -
Analysis 6.4

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 4 Dizziness.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 5 Somnolence.
Figuras y tablas -
Analysis 6.5

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 5 Somnolence.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 6 Adverse event leading to cessation.
Figuras y tablas -
Analysis 6.6

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 6 Adverse event leading to cessation.

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 7 Serious adverse event.
Figuras y tablas -
Analysis 6.7

Comparison 6 Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia, Outcome 7 Serious adverse event.

Summary of findings for the main comparison. Duloxetine for the treatment of painful diabetic neuropathy

Duloxetine for painful diabetic neuropathy

Patient or population: patients with painful neuropathy or chronic pain from diabetic peripheral neuropathy
Settings: primary and secondary care
Intervention: duloxetine

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Duloxetine

Number of patients with ≥ 50% improvement of pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert score

Follow‐up: 8 to 12 weeks

257 per 1000

445 per 1000
(370 to 535)

RR 1.73
(1.44 to 2.08)

908
(4 studies)

⊕⊕⊕⊝
moderate1

NNTB for ≥ 50% reduction in pain at 60 mg daily: 5 (95% CI 4 to 7)

Mean improvement in pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert score

Scale from: 0 to 10
Follow‐up: 8 to 12 weeks

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 60 mg daily in the control groups was
‐1.65 units

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 60 mg daily in the intervention groups was
0.96 lower
(1.26 to 0.65 lower)

722
(4 studies)

⊕⊕⊕⊝
moderate2

Number of patients with ≥ 30% improvement in pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert scale

Follow‐up: 8 to 12 weeks

411 per 1000

629 per 1000
(547 to 719)

RR 1.53
(1.33 to 1.75)

799
(4 studies)

⊕⊕⊕⊝
moderate1

NNTB for ≥ 30% reduction in pain at 60 mg duloxetine daily: 5 (95% CI 3 to 7)

Mean improvement in Patient Reported Global Impression of Change at 12 weeks or less

Duloxetine 60 mg daily
VAS

Scale from: 0 to 10
Follow‐up: 8 to 12 weeks

The mean mean improvement in patient reported global impression of improvement change at 12 weeks or less ‐ duloxetine 60 mg daily in the control groups was
‐3.06 units

The mean mean improvement in Patient Reported Global Impression of Improvement Change at 12 weeks or less ‐ duloxetine 60 mg daily in the intervention groups was
0.6 lower
(0.77 to 0.44 lower)

1018
(5 studies)

⊕⊕⊕⊝
moderate3

Adverse event leading to cessation

All neuropathic pain indications

Duloxetine 60 mg daily

56 per 1000

109 per 1000
(90 to 133)

RR 1.95
(1.6 to 2.37)

4837
(14 studies)

⊕⊕⊝⊝
low4

NNTH for duloxetine 60 mg daily, all indications, and all adverse effects leading to cessation: 18 (95% CI 13 to 30)

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; NNTB: number needed to treat for an additional beneficial outcome; NNTH: number needed to treat for an additional harmful outcome

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

1 Four trials, all company sponsored and performed but all trials pre‐registered on ClinicalTrials.gov have been published. No publication bias detected.
2 Two of four studies by company. Effect in Rowbotham nonsignificant, contributing some heterogeneity.
3 Five studies but wide CIs in the independent studies.
4 Variable quality of adverse event collection.

Figuras y tablas -
Summary of findings for the main comparison. Duloxetine for the treatment of painful diabetic neuropathy
Summary of findings 2. Duloxetine for the treatment of the chronic pain of fibromyalgia

Duloxetine for the chronic pain of fibromyalgia

Patient or population: patients with the chronic pain of fibromyalgia
Settings:
Intervention: duloxetine

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Duloxetine

Number with ≥ 50% improvement of pain at 12 weeks or less

Duloxetine 60 mg daily
11‐point Likert scale
Follow‐up: 8 to 12 weeks

233 per 1000

366 per 1000
(280 to 480)

RR 1.57
(1.2 to 2.06)

528
(2 studies)

⊕⊕⊝⊝
low1,2

NNTB for ≥ 50% improvement of pain at duloxetine 60 mg daily: 8 (95% CI 4 to 21)

Number with ≥ 30% improvement of pain at 12 weeks or less

Duloxetine 60 mg daily

Follow‐up: 8 to 12 weeks

347 per 1000

527 per 1000
(430 to 642)

RR 1.52
(1.24 to 1.85)

528
(2 studies)

⊕⊕⊝⊝
low1,2

NNTB for ≥ 30% improvement of pain at duloxetine 60 mg daily: NNT 6 (95% CI 3 to 12)

Mean improvement in the Patient Reported Global Impression of Change at completion of trial

Duloxetine 60 mg daily
VAS

Scale from: 0 to 10
Follow‐up: 12 weeks

The mean mean improvement in the patient reported global impression of change at completion of trial ‐ duloxetine 60 mg daily in the control groups was
3.52 units

The mean mean improvement in the patient reported global impression of change at completion of trial ‐ duloxetine 60 mg daily in the intervention groups was
0.45 lower
(0.73 to 0.18 lower)

519
(2 studies)

⊕⊕⊝⊝
low1,2

Mean improvement in pain at 12 weeks or less

Duloxetine 120 mg daily

LikertScale from: 0 to 10
Follow‐up: 12 weeks

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 120 mg daily in the control groups was
‐1.5

The mean mean improvement in pain at 12 weeks or less ‐ duloxetine 120 mg daily in the intervention groups was
0.8 lower
(1.35 to 0.25 lower)

507
(1 study)

⊕⊕⊕⊝
moderate1

Adverse events

See comment

See comment

See comment

See comment

See pooled adverse events in 'Summary of findings' table 1

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; NNTB: number needed to treat for an additional beneficial outcome

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

1 Substantial dropouts from all trials inform the outcomes.
2 Mostly female in some trials, all female in others.

Figuras y tablas -
Summary of findings 2. Duloxetine for the treatment of the chronic pain of fibromyalgia
Summary of findings 3. Duloxetine for the treatment of pain in major depressive disorder

Duloxetine for pain in major depressive disorder

Patient or population: patients with pain in major depressive disorder
Settings:
Intervention: duloxetine

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Duloxetine

Number with ≥ 50% pain relief at 12 weeks or less
Follow‐up: 12 weeks

360 per 1000

493 per 1000
(428 to 572)

RR 1.37
(1.19 to 1.59)

1023
(2 studies)

⊕⊕⊕⊝
moderate1

NNTB for ≥ 50% pain relief at < 12 weeks 60 mg duloxetine daily: 8 (95% CI 5 to 14)

Number with ≥ 30% pain relief at 12 weeks or less

467 per 1000

593 per 1000
(537 to 654)

RR 1.27
(1.15 to 1.4)

1359
(3 studies)

⊕⊕⊝⊝
low1,2

NNTB for ≥ 30% pain relief at < 12 weeks 60 mg duloxetine: 8 (95% CI 4‐ to 14)

Mean improvement in pain at 12 weeks or less
Visual analogue scale. Scale from: 0 to 10.
Follow‐up: 12 weeks

The mean mean improvement in pain at 12 weeks or less in the control groups was
1.23

The mean mean improvement in pain at 12 weeks or less in the intervention groups was
0.55 lower
(0.75 to 0.35 lower)

1359
(3 studies)

⊕⊕⊝⊝
low1,2

Mean improvement in Patient Reported Global Impression of Change at 12 weeks or less

See comment

See comment

Not estimable

See comment

Outcome not measured

Adverse events

See comment

See comment

Not estimable

See comment

See pooled adverse events in 'Summary of findings' table 1

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio; NNTB: number needed to treat for an additional beneficial outcome

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

1 Mixed causes for pain, not necessarily neuropathic.
2 Substantial dropouts partially accounted for by last observation carried forward and statistical manipulation.

Figuras y tablas -
Summary of findings 3. Duloxetine for the treatment of pain in major depressive disorder
Comparison 1. Duloxetine versus placebo in the treatment of painful diabetic neuropathy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Duloxetine 20 mg daily

1

213

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.98, 2.09]

1.2 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Random, 95% CI)

1.91 [1.26, 2.87]

1.3 Duloxetine 60 mg daily

4

908

Risk Ratio (M‐H, Random, 95% CI)

1.73 [1.44, 2.08]

1.4 Duloxetine 120 mg daily

4

870

Risk Ratio (M‐H, Random, 95% CI)

1.46 [1.08, 1.97]

1.5 All doses

5

1655

Risk Ratio (M‐H, Random, 95% CI)

1.53 [1.21, 1.92]

2 Mean improvement in pain at 12 weeks or less Show forest plot

5

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

2.1 Duloxetine 20 mg daily

1

179

Mean Difference (IV, Fixed, 95% CI)

‐0.45 [‐1.05, 0.15]

2.2 Duloxetine 60 mg daily

4

722

Mean Difference (IV, Fixed, 95% CI)

‐0.96 [‐1.26, ‐0.65]

2.3 Duloxetine 120 mg daily

4

828

Mean Difference (IV, Fixed, 95% CI)

‐0.93 [‐1.21, ‐0.65]

3 Number of participants with ≥ 30% improvement in pain at 12 weeks or less Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

1.57 [1.18, 2.07]

3.2 Duloxetine 60 mg daily

4

799

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [1.33, 1.75]

3.3 Duloxetine 120 mg daily

3

659

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [1.21, 1.58]

3.4 All doses

4

1220

Risk Ratio (M‐H, Fixed, 95% CI)

1.45 [1.30, 1.63]

4 Mean improvement in SF‐36 Physical Subscore at 12 weeks or less Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 Duloxetine 20 mg daily

1

200

Mean Difference (IV, Random, 95% CI)

‐0.27 [‐2.42, 1.88]

4.2 Duloxetine 60 mg daily

3

514

Mean Difference (IV, Random, 95% CI)

2.65 [1.38, 3.92]

4.3 Duloxetine 120 mg daily

2

409

Mean Difference (IV, Random, 95% CI)

2.80 [1.04, 4.55]

5 Mean improvement in SF‐36 Mental Subscore at 12 weeks or less Show forest plot

3

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

5.1 Duloxetine 20 mg daily

1

200

Mean Difference (IV, Fixed, 95% CI)

1.11 [‐0.98, 3.20]

5.2 Duloxetine 60 mg daily

3

514

Mean Difference (IV, Fixed, 95% CI)

1.08 [‐0.32, 2.48]

5.3 Duloxetine 120 mg daily

2

409

Mean Difference (IV, Fixed, 95% CI)

2.23 [0.69, 3.77]

6 Mean improvement in SF‐36 Bodily Pain Subscore at 12 weeks or less Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

6.1 Duloxetine 20 mg daily

1

209

Mean Difference (IV, Fixed, 95% CI)

2.90 [‐2.37, 8.17]

6.2 Duloxetine 60 mg daily

2

421

Mean Difference (IV, Fixed, 95% CI)

5.58 [1.74, 9.42]

6.3 Duloxetine 120 mg daily

2

420

Mean Difference (IV, Fixed, 95% CI)

8.19 [4.33, 12.05]

7 Mean improvement in Patient Reported Global Impression of Improvement at 12 weeks or less Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

7.1 Duloxetine 20 mg daily

1

219

Mean Difference (IV, Random, 95% CI)

‐0.23 [‐0.56, 0.10]

7.2 Duloxetine 40 mg daily

1

252

Mean Difference (IV, Random, 95% CI)

‐0.65 [‐1.01, ‐0.29]

7.3 Duloxetine 60 mg daily

5

1018

Mean Difference (IV, Random, 95% CI)

‐0.60 [‐0.77, ‐0.44]

7.4 Duloxetine 120 mg daily

4

870

Mean Difference (IV, Random, 95% CI)

‐0.54 [‐0.73, ‐0.35]

8 Mean improvement in BPI Severity ‐ average pain at 12 weeks or less Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

8.1 Duloxetine 60 mg daily

2

433

Mean Difference (IV, Random, 95% CI)

‐0.97 [‐1.38, ‐0.57]

8.2 Duloxetine 120 mg daily

2

428

Mean Difference (IV, Random, 95% CI)

‐1.16 [‐1.91, ‐0.41]

9 Mean improvement in pain at rest (night pain) at 12 weeks or less Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

9.1 Duloxetine 20 mg daily

1

222

Mean Difference (IV, Random, 95% CI)

‐0.28 [‐0.90, 0.34]

9.2 Duloxetine 60 mg daily

3

664

Mean Difference (IV, Random, 95% CI)

‐0.92 [‐1.27, ‐0.57]

9.3 Duloxetine 120 mg daily

3

664

Mean Difference (IV, Random, 95% CI)

‐1.10 [‐1.45, ‐0.75]

Figuras y tablas -
Comparison 1. Duloxetine versus placebo in the treatment of painful diabetic neuropathy
Comparison 2. Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with ≥ 50% improvement in pain at 12 weeks or less Show forest plot

1

804

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [1.19, 1.80]

2 Mean improvement in pain at 12 weeks or less Show forest plot

1

804

Mean Difference (IV, Fixed, 95% CI)

‐0.62 [‐0.92, ‐0.32]

3 Number improved ≥ 30% at 12 weeks or less Show forest plot

1

804

Risk Ratio (M‐H, Fixed, 95% CI)

1.42 [1.20, 1.68]

Figuras y tablas -
Comparison 2. Duloxetine versus pregabalin in the treatment of painful diabetic neuropathy
Comparison 3. Duloxetine versus placebo in the treatment of fibromyalgia

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with ≥ 50% improvement of pain at 12 weeks or less Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

1.1 Duloxetine 20 mg daily

1

223

Risk Ratio (M‐H, Fixed, 95% CI)

1.39 [0.91, 2.14]

1.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.75, 1.35]

1.3 Duloxetine 60 mg daily

2

528

Risk Ratio (M‐H, Fixed, 95% CI)

1.57 [1.20, 2.06]

1.4 Duloxetine 120 mg daily

4

1234

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [1.40, 2.03]

1.5 All doses

5

1887

Risk Ratio (M‐H, Fixed, 95% CI)

1.50 [1.29, 1.75]

2 Number of participants with ≥ 50% improvement of pain at more than 12 weeks Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2.1 Duloxetine 60 mg daily

1

373

Risk Ratio (M‐H, Fixed, 95% CI)

1.58 [1.10, 2.27]

2.2 Duloxetine 120 mg daily

2

616

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [1.07, 1.79]

2.3 Duloxetine all doses

2

845

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [1.09, 1.79]

3 Number of participants with ≥ 30% improvement of pain at 12 weeks or less Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Duloxetine 20 mg daily

1

223

Risk Ratio (M‐H, Fixed, 95% CI)

1.30 [0.94, 1.79]

3.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.89, 1.45]

3.3 Duloxetine 60 mg daily

2

528

Risk Ratio (M‐H, Fixed, 95% CI)

1.52 [1.24, 1.85]

3.4 Duloxetine 120 mg daily

3

1020

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [1.26, 1.69]

3.5 All doses

4

1673

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [1.22, 1.56]

4 Mean improvement in pain at 12 weeks or less Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

4.1 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Fixed, 95% CI)

‐0.31 [‐0.86, 0.24]

4.2 Duloxetine 120 mg daily

1

507

Mean Difference (IV, Fixed, 95% CI)

‐0.80 [‐1.35, ‐0.25]

5 Mean improvement in the SF‐36 mental component summary subscore Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

5.1 Duloxetine 20 mg daily

1

223

Mean Difference (IV, Random, 95% CI)

0.81 [‐2.37, 3.99]

5.2 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Random, 95% CI)

2.69 [0.31, 5.07]

5.3 Duloxetine 60 mg daily

2

515

Mean Difference (IV, Random, 95% CI)

3.31 [0.59, 6.02]

5.4 Duloxetine 120 mg daily

5

1531

Mean Difference (IV, Random, 95% CI)

4.22 [2.43, 6.02]

6 Mean improvement in the SF‐36 physical component summary subscore Show forest plot

6

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

6.1 Duloxetine 20 mg daily

1

223

Mean Difference (IV, Fixed, 95% CI)

0.81 [‐1.92, 3.54]

6.2 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Fixed, 95% CI)

0.84 [‐1.17, 2.85]

6.3 Duloxetine 60 mg daily

2

515

Mean Difference (IV, Fixed, 95% CI)

1.28 [‐0.33, 2.89]

6.4 Duloxetine 120 mg daily

5

1531

Mean Difference (IV, Fixed, 95% CI)

2.13 [0.95, 3.30]

7 Mean improvement in the SF‐36 Bodily Pain Subscore Show forest plot

4

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

7.1 Duloxetine 60 mg daily

1

221

Mean Difference (IV, Fixed, 95% CI)

8.2 [3.20, 13.20]

7.2 Duloxetine 120 mg daily

4

1243

Mean Difference (IV, Fixed, 95% CI)

5.96 [3.76, 8.16]

8 Mean improvement in the Patient reported Global Impression of Change at completion of trial Show forest plot

4

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

8.1 Duloxetine 20 mg daily

1

223

Mean Difference (IV, Fixed, 95% CI)

‐0.54 [‐0.96, ‐0.12]

8.2 Duloxetine 30 mg daily

1

308

Mean Difference (IV, Fixed, 95% CI)

‐0.38 [‐0.71, ‐0.05]

8.3 Duloxetine 60 mg daily

2

519

Mean Difference (IV, Fixed, 95% CI)

‐0.45 [‐0.73, ‐0.18]

8.4 Duloxetine 120 mg daily

3

826

Mean Difference (IV, Fixed, 95% CI)

‐0.44 [‐0.66, ‐0.23]

Figuras y tablas -
Comparison 3. Duloxetine versus placebo in the treatment of fibromyalgia
Comparison 4. Duloxetine versus placebo for the treatment of pain in major depressive disorder

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of participants with > 50% pain relief at 12 weeks or less Show forest plot

2

1023

Risk Ratio (M‐H, Fixed, 95% CI)

1.37 [1.19, 1.59]

2 Participants with > 30% pain relief at 12 weeks or less Show forest plot

3

1359

Risk Ratio (M‐H, Fixed, 95% CI)

1.27 [1.15, 1.40]

3 Mean improvement in pain at 12 weeks or less Show forest plot

3

1359

Mean Difference (IV, Fixed, 95% CI)

‐0.55 [‐0.75, ‐0.35]

Figuras y tablas -
Comparison 4. Duloxetine versus placebo for the treatment of pain in major depressive disorder
Comparison 5. Duloxetine versus placebo in the treatment of central neuropathic pain

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mean improvement in pain at 12 weeks or less Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

‐1.0 [‐2.05, 0.05]

2 Mean improvement in SF‐36 Physical Subscore Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

2.0 [‐12.72, 16.72]

3 Mean improvement in the SF‐36 Mental Subscore at 12 weeks Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

4.0 [‐6.75, 14.75]

4 Mean improvement in the SF‐36 Bodily Pain Subscore Show forest plot

1

48

Mean Difference (IV, Fixed, 95% CI)

8.0 [‐0.81, 16.81]

5 Number of participants improved on PGI‐I (better or very much better) Show forest plot

1

48

Risk Ratio (M‐H, Fixed, 95% CI)

2.75 [1.02, 7.44]

Figuras y tablas -
Comparison 5. Duloxetine versus placebo in the treatment of central neuropathic pain
Comparison 6. Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of participants with any adverse event Show forest plot

14

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

1.1 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.25 [1.03, 1.52]

1.2 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.01, 1.31]

1.3 Duloxetine 60 mg daily

13

4521

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.10, 1.20]

1.4 Duloxetine 120 mg daily

3

688

Risk Ratio (M‐H, Fixed, 95% CI)

1.19 [1.09, 1.30]

1.5 Duloxetine all doses

14

5258

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.11, 1.20]

2 Nausea Show forest plot

13

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Random, 95% CI)

1.45 [0.71, 3.00]

2.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Random, 95% CI)

5.43 [2.34, 12.58]

2.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Random, 95% CI)

6.55 [1.85, 23.17]

2.4 Duloxetine 60 mg daily

11

3642

Risk Ratio (M‐H, Random, 95% CI)

2.61 [2.14, 3.18]

2.5 Duloxetine 120 mg daily

4

787

Risk Ratio (M‐H, Random, 95% CI)

2.89 [2.06, 4.04]

3 Dry mouth Show forest plot

7

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.30, 2.47]

3.2 Duloxetine 40 mg daily

0

0

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.3 Duloxetine 60 mg daily

6

2004

Risk Ratio (M‐H, Fixed, 95% CI)

2.63 [1.89, 3.67]

3.4 Duloxetine 120 mg daily

3

567

Risk Ratio (M‐H, Fixed, 95% CI)

3.40 [1.94, 5.96]

4 Dizziness Show forest plot

9

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

4.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.33, 2.33]

4.2 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

5.89 [1.22, 28.58]

4.3 Duloxetine 60 mg daily

8

2257

Risk Ratio (M‐H, Fixed, 95% CI)

1.84 [1.35, 2.51]

4.4 Duloxetine 120 mg daily

4

787

Risk Ratio (M‐H, Fixed, 95% CI)

2.44 [1.55, 3.83]

5 Somnolence Show forest plot

10

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

5.1 Duloxetine 20 mg daily

1

230

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.41, 2.43]

5.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

2.22 [0.70, 7.06]

5.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

2.25 [1.15, 4.38]

5.4 Duloxetine 60 mg daily

8

2678

Risk Ratio (M‐H, Fixed, 95% CI)

2.94 [2.17, 3.97]

5.5 Duloxetine 120 mg daily

4

787

Risk Ratio (M‐H, Fixed, 95% CI)

4.76 [2.93, 7.74]

6 Adverse event leading to cessation Show forest plot

17

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

6.1 Duloxetine 20 mg daily

2

453

Risk Ratio (M‐H, Fixed, 95% CI)

1.37 [0.78, 2.39]

6.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

1.54 [0.69, 3.44]

6.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

1.96 [0.81, 4.77]

6.4 Duloxetine 60 mg daily

14

4837

Risk Ratio (M‐H, Fixed, 95% CI)

1.95 [1.60, 2.37]

6.5 Duloxetine 120 mg daily

7

1462

Risk Ratio (M‐H, Fixed, 95% CI)

2.30 [1.74, 3.04]

6.6 All doses

17

6285

Risk Ratio (M‐H, Fixed, 95% CI)

1.99 [1.67, 2.37]

7 Serious adverse event Show forest plot

16

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

7.1 Duloxetine 20 mg daily

0

0

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Duloxetine 30 mg daily

1

308

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.01, 8.02]

7.3 Duloxetine 40 mg daily

1

252

Risk Ratio (M‐H, Fixed, 95% CI)

2.95 [0.50, 17.30]

7.4 Duloxetine 60 mg daily

14

4842

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.60, 1.32]

7.5 Duloxetine 120 mg daily

6

1257

Risk Ratio (M‐H, Fixed, 95% CI)

0.59 [0.25, 1.35]

7.6 All doses

14

4976

Risk Ratio (M‐H, Fixed, 95% CI)

0.81 [0.53, 1.25]

Figuras y tablas -
Comparison 6. Duloxetine versus placebo: adverse events during first 12 weeks of treatment for painful neuropathy or fibromyalgia