Télécharger au format pdf ou txt
Télécharger au format pdf ou txt
Vous êtes sur la page 1sur 28

revue neurologique 176 (2020) 325–352

Available online at

ScienceDirect
www.sciencedirect.com

Practice guidelines

Pharmacological and non-pharmacological


treatments for neuropathic pain: Systematic review
and French recommendations

X. Moisset a,b,*, D. Bouhassira c,d, J. Avez Couturier e, H. Alchaar f,


S. Conradi g, M.H. Delmotte h, M. Lanteri-Minet a,i, J.P. Lefaucheur j,k,
G. Mick l, V. Piano m, G. Pickering a,n, E. Piquet i, C. Regis o, E. Salvat p,
N. Attal c,d
a
Université Clermont Auvergne, Inserm, Neuro-Dol, 63000 Clermont-Ferrand, France
b
CHU de Clermont-Ferrand, 63000 Clermont-Ferrand, France
c
INSERM U987, CETD, Ambroise-Paré Hospital, AP–HP, Boulogne-Billancourt, France
d
Université Versailles – Saint-Quentin-en-Yvelines, Versailles, France
e
Service de Neuropédiatrie, Consultation Douleur Enfant, CIC-IT 1403, CHU de Lille, Lille, France
f
73, boulevard de Cimiez, Nice, France
g
CETD, CHRU de Nancy, Vandœuvre-lès-Nancy, France
h
GHU, Paris site Ste-Anne, Structure Douleurs, 1, rue Cabanis, Paris 14, France
i
Département d’Évaluation et Traitement de la Douleur, Centre Hospitalier Universitaire (CHU) de Nice, Fédération
Hospitalo-Universitaire InovPain, Université Côte d’Azur, Nice, France
j
EA 4391, Faculté de Médecine, Université Paris Est Créteil, Créteil, France
k
Service de Physiologie, Explorations Fonctionnelles, Hôpital Henri-Mondor, Assistance publique–Hôpitaux de Paris,
Créteil, France
l
Centre d’Évaluation et Traitement de la Douleur du Voironnais, Centre Hospitalier de Voiron, Laboratoire P2S,
Université de Lyon, Lyon, France
m
Centre Hospitalier de Draguignan, Service Algologie 4e, route de Montferrat, 83007 Draguignan cedex, France
n
Clinical Pharmacology Department, CPC/CIC Inserm 1405, University Hospital CHU, Clermont-Ferrand, France
o
CETD, CHU Montpellier, Montpellier, France
p
Centre d’Évaluation et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Université de
Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique,
Strasbourg, France

* Corresponding author at: Service de Neurologie, 58, rue Montalembert, 63000 Clermont-Ferrand, France.
E-mail address: xavier.moisset@gmail.com (X. Moisset).
https://doi.org/10.1016/j.neurol.2020.01.361
0035-3787/# 2020 Elsevier Masson SAS. All rights reserved.
326 revue neurologique 176 (2020) 325–352

info article abstract

Article history: Neuropathic pain remains a significant unmet medical need. Several recommendations
Received 23 November 2019 have recently been proposed concerning pharmacotherapy, neurostimulation techniques
Accepted 7 January 2020 and interventional management, but no comprehensive guideline encompassing all these
Available online 7 April 2020 treatments has yet been issued. We performed a systematic review of pharmacotherapy,
neurostimulation, surgery, psychotherapies and other types of therapy for peripheral or
Keywords: central neuropathic pain, based on studies published in peer-reviewed journals before
Neuropathic pain January 2018. The main inclusion criteria were chronic neuropathic pain for at least three
Systematic review months, a randomized controlled methodology, at least three weeks of follow-up, at least 10
Randomized controlled trials patients per group, and a double-blind design for drug therapy. Based on the GRADE system,
Recommendations we provide weak-to-strong recommendations for use and proposal as a first-line treatment
Pharmacotherapy for SNRIs (duloxetine and venlafaxine), gabapentin and tricyclic antidepressants and, for
Neurostimulation topical lidocaine and transcutaneous electrical nerve stimulation specifically for peripheral
Surgery neuropathic pain; a weak recommendation for use and proposal as a second-line treatment
Psychotherapy for pregabalin, tramadol, combination therapy (antidepressant combined with gabapenti-
noids), and for high-concentration capsaicin patches and botulinum toxin A specifically for
peripheral neuropathic pain; a weak recommendation for use and proposal as a third-line
treatment for high-frequency rTMS of the motor cortex, spinal cord stimulation (failed back
surgery syndrome and painful diabetic polyneuropathy) and strong opioids (in the absence
of an alternative). Psychotherapy (cognitive behavioral therapy and mindfulness) is recom-
mended as a second-line therapy, as an add-on to other therapies. An algorithm encom-
passing all the recommended treatments is proposed.
# 2020 Elsevier Masson SAS. All rights reserved.

1. Introduction patients or elderly individuals with neuropathic pain. In


France, neuropathic pain management in children is currently
Neuropathic pain, caused by a lesion or disease affecting the governed by the guidelines published by the French regulatory
somatosensory nervous system, has a considerable impact on authorities for drugs in 2009 [16], which are mostly based on
quality of life and is associated with a high economic burden adult guidelines and expert consensus.
on the individual and society [1–4]. Here, we aimed to propose new French recommendations
Epidemiological surveys have shown that many patients encompassing all the available therapeutic options for
with neuropathic pain do not receive appropriate treatment neuropathic pain. We used the Grading of Recommendations
for their pain [2,5–9]. This may be due to a lack of diagnostic Assessment, Development, and Evaluation (GRADE) system
accuracy, insufficient understanding of the pathophysiologi- [17,18] to propose a comprehensive therapeutic algorithm
cal mechanisms of pain, relatively ineffective drugs and encompassing all medical, psychological and surgical thera-
insufficient knowledge of the effective drugs and their pies for neuropathic pain. We performed a systematic review
appropriate use in clinical practice. Other non-pharmacolo- of randomized comparative or placebo-controlled trials with a
gical treatments, including noninvasive neurostimulation double-blind design, but we extended our search to single-
techniques, psychotherapy, complementary therapy and blind or open-label trials for treatments for which double
invasive approaches, are increasingly being offered to patients blinding is difficult or impossible, such as psychotherapy and
with neuropathic pain, most of whom are treated with a surgery. For topical and oral pharmacotherapies, we searched
combination of therapeutic approaches in routine clinical for articles published since June 2013, making use of the most
practice [10,11]. However, recommendations for the treatment recent systematic review and meta-analysis of pharmaco-
of neuropathic pain have always been restricted to a specific therapy jointly directed by one senior member of our group
type of treatment, such as pharmacotherapy or neurostimula- [13] for older articles, and, for other therapies, we searched for
tion. Thus, for example, the previous French guidelines for articles published since 1966 (first studies published in
neuropathic pain treatment published in 2010 [12] focused MEDLINE). Disease-modifying treatments and specific treat-
principally on pharmacotherapy. Since the publication of ments for trigeminal neuralgia or erythromelalgia are beyond
these guidelines, international evidence-based recommenda- the scope of this work [19].
tions have been issued for pharmacotherapy [13], interven-
tional management [14] and central neurostimulation [15]. No
evidence-based recommendations encompassing all the 2. Methods
available therapeutic approaches have yet been proposed
for neuropathic pain. Furthermore, most of the previous This systematic review (PROSPERO CRD42019140749) was
recommendations considered adults, but not pediatric prepared under the auspices of the International Association
revue neurologique 176 (2020) 325–352 327

for the Study of Pain (IASP) French Chapter (‘‘Société française facial pain were not included, because they do not fulfill the
pour l’étude et le traitement de la douleur’’, SFETD) and the current definition of neuropathic pain. Trigeminal neuralgia
French Society of Neurology (‘‘Société française de neurolo- was excluded because of generally distinct response to drug
gie’’, SFN) and supported by ‘‘Association nationale pédago- treatment.
gique des enseignants en thérapeutique’’, APNET (‘‘French The main inclusion criteria common to all treatment
college of therapeutic teachers’’). We followed the 23-item groups were: chronic neuropathic pain (for at least 3 months);
Appraisal of Guidelines for Research and Evaluation (AGREE II) single (e.g. iv drugs) or repeated administrations of treatment;
Instrument for the development and reporting of recommen- randomized design; comparison with a control group (placebo
dations [20]. Details of the search criteria and methods can be or comparator) over a follow-up period of at least three weeks
found in Appendix 1. The guidelines proposed by the French (i.e. at least three weeks of assessment of the efficacy of active
health authority (‘‘Haute Autorité de santé’’) were applied treatment and of comparator in the case of crossover trials); at
(‘‘Recommandations pour la pratique clinique’’, [21]). The least 10 patients per group; pain assessed as a primary
steering group consisted of 15 healthcare professionals outcome; studies published as full reports; any treatment for
(Appendix 2). A reading committee of 32 healthcare pro- neuropathic pain, regardless of label, available for use in
fessionals from the SFETD, SFN, other independent reviewers France. We anticipated significant heterogeneity in trial
and patients (‘‘Association francophone pour vaincre les quality. We therefore required a double-blind design for
douleurs’’; AFVD) was set up and scored the proposals studies of pharmacotherapy, radiofrequency and noninvasive
(Appendix 2). This work was supported by a grant from SFN. brain neurostimulation, but we extended our search to single-
blind or open-label controlled studies for TENS, invasive
2.1. Procedures neurostimulation, surgery, psychotherapy and hypnosis,
because double-blind studies were rare for these treatments
The systematic literature review was performed according to (mostly due to the difficulty achieving blinding for these
the Preferred Reporting Items for Systematic Reviews and treatments). For example, TENS and conventional spinal cord
Meta-Analyses (PRISMA) statements [22]. We used a standar- stimulation induce paresthesia in the painful area, so it is
dized review and data extraction protocol (Appendix 3). difficult to blind the patient to the treatment group.
Randomized controlled trials (RCTs) published as full reports
in peer-reviewed journals until January 18, 2018 were 2.2. Evidence summary and reporting
identified with PubMed/MEDLINE and Embase. We searched
for studies on oral or topical drug treatments published since We used the GRADE system (Appendix 4) to assess the risk of
June 2013. For studies published from 1966 to June 2013, we bias for individual studies and groups of studies and to
referred to the most recent systematic review (SR) and meta- propose recommendations.
analysis on this subject, which was published in 2015 [13],
used the GRADE system, had similar inclusion criteria and was 2.2.1. Risk of bias in individual trials
jointly directed by the senior author of this study (NA). For all Two review authors for each treatment independently
other treatments, we searched for articles published since assessed the risk of bias for each study using the GRADE
1966 (these treatments included oral ketamine, intrathecal system for individual studies. The methods are described in
therapy, IV drugs, nerve blocks, neurostimulation, surgery or detail in Appendix 4. We assessed the following for each study:
psychotherapy). Additional articles were identified from
published reviews (especially Cochrane reviews), the refe-
rence lists of the selected papers and additional searches of  random sequence generation (checking for possible selec-
the same databases for publications appearing up to August tion bias) and allocation concealment;
2019.  blinding of participants (checking for possible performance
The target population was patients of any age, including bias);
children, with neuropathic pain according to the IASP  blinding of personnel;
definition (pain caused by a lesion or disease of the  incomplete outcome data (checking for possible attrition
somatosensory nervous system), consistent with that used bias due to the amount, nature, and handling of incomplete
for the 2015 meta-analysis [13]. The neuropathic pain outcome data);
conditions considered included postherpetic neuralgia, dia-  statistical methods;
betic and non-diabetic painful polyneuropathy, post-amputa-  sample size calculation;
tion pain, post-traumatic/postsurgical neuropathic pain  selective reporting;
including plexus avulsion and complex regional pain syn-  carryover effect, evaluated for crossover studies and noted
drome type II (post-traumatic/postsurgical neuropathic pain), as ‘‘not applicable’’ for parallel group studies;
central post-stroke pain, spinal cord injury (SCI) pain, and  the presence of any other bias evaluated separately,
multiple sclerosis-associated pain. Multiple etiologies of including small sample size or poor description of neuropa-
neuropathic pain were considered. Neuropathic pain associa- thic pain definition in particular.
ted with nociceptive components (e.g. cancer neuropathic
pain and radiculopathy) was included provided that the The overall quality of the study was considered ‘‘very high’’
neuropathic pain was considered separately. Conditions such if no bias was noted, ‘‘high’’ if there were only one or two
as complex regional pain syndrome type I, low back pain biases, ‘‘moderate’’ in the presence of three or four biases and
without radicular pain, fibromyalgia, and persistent idiopathic ‘‘low to very low’’ if there was five or more biases. This
328 revue neurologique 176 (2020) 325–352

classification differs slightly from that used in previous the treatment, and was based on expert consensus for
systematic reviews of neuropathic pain treatments [13,15] in conditions for which high-quality trials were lacking, for
which the ‘‘low’’ and ‘‘very low’’ levels were generally elderly or pediatric populations, for example. These recom-
considered separately. Indeed, it was often difficult to mendations do not take treatment costs into account because
distinguish between trials of low and very low-quality, and these costs may differ from country to country and may
we felt that pooling these two levels together would be change over time. For the same reasons, the indications
unlikely to affect our recommendations (we did not recom- proposed by regulatory agencies for use in neuropathic pain
mend treatments for which there was a low or very low level of were not taken into account.
evidence for use).

2.2.2. Risk of bias across studies 3. Results


Heterogeneity, imprecision, small effect size and indirectness
were evaluated with the GRADE system for a group of studies 3.1. Study selection and characteristics
(see Appendix 4 for detailed methods). These factors led to the
downgrading (when present) or upgrading (for the opposite The results of the database and registry search are shown in
situation, e.g. large effect sizes) of the quality of evidence for a Fig. 1A for pharmacological treatments and Fig. 1B for non-
group of studies. The risk of publication bias was not pharmacological therapies. We included a total of 131
specifically evaluated because no systematic search for published articles in the quantitative analysis. The characte-
unpublished trials was performed. ristics of the studies concerned are summarized in
The GRADE system was also used to propose recommen- Appendix 5.
dations based on a group of RCTs pertaining to the same drug Eligible studies investigated:
class or technique. Final recommendations, based on the
GRADE system, were rated as strongly in favor of the
treatment, weakly in favor of the treatment, strongly or  pharmacological treatments: antidepressants, antiepileptic
weakly against the treatment, or inconclusive. These levels of agents, opioids, topical agents (mostly lidocaine and cap-
recommendations were based on the balance between efficacy saicin), subcutaneous botulinum toxin type A (BTX-A),
and adverse effects, patient satisfaction (if data were combination therapy, cannabinoids, oral ketamine, intra-
available), and on prior evidence provided by the most recent venous drugs (e.g. ketamine, lidocaine, opioids, other),
systematic review of oral and topical pharmacotherapy [13]. intrathecal drugs (e.g. morphine, clonidine, local anesthe-
tics, ziconotide), nerve blocks, newer drugs;
2.3. Outcome measures  noninvasive peripheral stimulation (including TENS), inva-
sive peripheral stimulation, invasive and noninvasive
We did not conduct a meta-analysis for this systematic study. central neurostimulation, pulsed radiofrequency and other
Instead, we report the number needed to treat (NNT) for a 50% surgical procedures;
decrease in pain intensity (or alternatively, a 30% decrease in  psychotherapies and other non-pharmacological treat-
pain intensity or at least moderate pain relief) and/or the effect ments not listed above.
size for individual trials, to obtain an estimate of the
magnitude of effects on the primary outcome (pain intensity), Overall, 47% (n = 61) of all the trials considered concerned
if such data were available. In tables (Appendix 5), efficacy was diabetic painful polyneuropathy (DPN) or postherpetic neural-
assessed on the basis of effect size, absolute difference gia (PHN); 11% (n = 14) concerned central pain, and 42%
between active and placebo or sham, or NNT. Effects on other concerned other neuropathic conditions (traumatic, radicular,
outcome measures such as neuropathic symptoms and signs, HIV, chemotherapy-induced, mixed etiologies). Few studies
sleep, quality of life, comorbidities, and Clinical Global lasted longer than 12 weeks, and the longest study duration
Impression of Change score, were reported when relevant for the trials included was 24 weeks.
(for example, if no efficacy was observed for the primary
outcome measure), but were considered secondary. Serious 3.2. Oral and topical drug therapy
and common (> 10% incidence) reported adverse events and
dropouts due to adverse effects were recorded (Appendix 5). We used the results of the previous systematic review (SR)
[13], which considered studies published from 1966 to June
2.4. Therapeutic algorithm 2013, and new studies published after this date in the
assessment of oral and topical drug treatments (except for
We developed a therapeutic algorithm for first-, second- and oral ketamine).
third-line treatments for all the pharmacological and non-
pharmacological treatments available in France, with a 3.2.1. Antidepressants
‘‘strong’’ or ‘‘weak’’ recommendation in favor of use based We identified 18 low- to high-quality placebo-controlled trials
on the GRADE system applied to all studies published since of tricyclic antidepressants (TCAs) (16 positive) in neuropathic
1966 (studies of oral and pharmacotherapy published before pain published before June 2013, with a final moderate quality
June 2013 being assessed in the meta-analysis of Finnerup of evidence. TCAs (nortriptyline and imipramine) were studied
et al., 2015). The level of recommendation in the algorithm also in two more recent high-quality RCTs of combination therapy
took into account ease of use and long-term experience with (125 patients treated) [23,24]. Each of the two drugs from the
revue neurologique 176 (2020) 325–352 329

Fig. 1 – A. PRISMA flowchart for pharmacological treatments. B. PRISMA flowchart for non-pharmacological interventions.
Studies comparing different treatments can be included more than once in the flowchart. ADs: antidepressants; AEDs:
antiepileptic drugs; Canna: cannabinoids; keta: ketamine; lido: lidocaine; BTX: botulinum toxin; IV: intravenous; IT:
intrathecal. TENS: transcutaneous electrical nerve stimulation; rTMS: repetitive transcranial magnetic stimulation; tDCS:
transcutaneous direct current stimulation; SCS: spinal cord stimulation; EMCS: epidural motor cortex stimulation; PRF:
pulsed radiofrequency; CBT: cognitive behavioral therapy; DRG stim: dorsal root ganglion stimulation.

combination was superior to placebo when administered as a Nine placebo-controlled trials of duloxetine (seven yielding
monotherapy. The adverse effects reported included central positive results) and four placebo-controlled trials of venlafa-
and anticholinergic effects. The final quality of evidence was xine ER (two yielding positive results) in neuropathic pain were
high. published before June 2013, with a high-quality of evidence.
330 revue neurologique 176 (2020) 325–352

We identified five more recent RCTs of duloxetine (60–120 mg/ studies gave results that were close to significance (588 patients
day; 565 with active treatment and 566 controls including 415 treated): in one, the results became significant if one patient
placebos) including three high-quality placebo-controlled trials was excluded [31] and in the other, a long-term trial (15 weeks),
[25–27] and two comparative trials versus nortriptyline (low- positive results were obtained at all earlier time points [32]. All
quality trial) or pregabalin (high-quality trial) [23,28]. Diabetic but one of the trials (the exception being a trial in which
painful neuropathy was the most frequent pain condition pregabalin was an active comparator) were characterized by
among the patients studied. The placebo-controlled trials very small effect sizes and/or large NNTs, with high rates of
yielded positive results for the primary outcome and for most placebo response. Potential confounding factors decreasing
secondary outcomes, including sleep, mood and quality of life, efficacy included the use of moderate doses of pregabalin in
with a small effect size in the two largest studies and a large most trials (300 mg/day, although this dose is similar to that in
effect size in a single-center study on lumbar radiculitis [27]. routine use), the inclusion of patients with refractory condi-
The two comparative studies concluded that duloxetine was tions [33] and large carryover effects [34,35]. One recent double-
non-inferior to nortriptyline or pregabalin. The most frequently blind study comparing pregabalin (150 to 300 mg) to gabapentin
reported adverse effects were nausea, loss of appetite, (1800 to 2400 mg) in chronic sciatica found that pregabalin was
somnolence, asthenia and dry mouth. One study in patients significantly less effective than gabapentin (difference from
with multiple sclerosis [25] found that discontinuation rates baseline: 1.72 for gabapentin versus 0.94 for pregabalin) [36].
were higher for duloxetine than for placebo. All these studies Adverse effects, mostly dizziness and drowsiness, were
were in favor of the use of duloxetine, with a high final quality common, and were significantly more frequent than for
of evidence. There was no recent RCT for venlafaxine. gabapentin in the comparative trial in patients with sciatica
(81% versus 19%) [36]. No severe side effects were reported.
These recent studies generally supported a lack of efficacy of
pregabalin, with a high final quality of evidence.
Recommendation: SNRIs (duloxetine and venlafaxine) Studies published before our search date for other anti-
and TCAs have a strong recommendation for use in epileptic drugs yielded negative or inconclusive results. One
neuropathic pain (high final quality of evidence), and recent single-center placebo-controlled trial assessed oxcar-
duloxetine should be preferred over venlafaxine (more bazepine in a population of patients with peripheral neuropa-
high-quality trials yielding positive results). TCAs should thic pain stratified for phenotype [37]; significant efficacy was
be monitored carefully if used at high doses (> 75 mg reported for the drug only in patients from the ‘‘irritable
daily) because of potential risks of hypotension and nociceptor’’ group (limited thermal deficit in the painful area);
cardiac toxicity. Lower doses should be preferred. patients with burning and paroxysmal pain (according to the
NPSI questionnaire) were also more responsive to the active
drug. However, this study had several biases (Appendix 5),
including a high dropout rate and potential unblinding (most
3.2.2. Antiepileptic drugs patients recognized the active treatment because of adverse
We identified 14 placebo-controlled trials of gabapentin (nine effects). One recent small placebo-controlled study in patients
yielding positive results) and six RCTs of gabapentin ER or with painful small-fiber neuropathy related to Nav1.7 muta-
enacarbil (four yielding positive results) published before June tion (n = 24) reported positive results for lacosamide [38]. The
2013, with a high final quality of evidence. Three more recent final quality of evidence was low.
high-quality double-blind trials evaluated the efficacy of gaba-
pentin or gabapentin enacarbil/ER against peripheral neuropa-
thic pain (132 patients treated). One comparative trial showed
gabapentin to be superior to pregabalin, with fewer side effects Recommendation: Gabapentin has a strong recommen-
(see the section on ‘‘pregabalin’’), another showed gabapentin dation for use in neuropathic pain (high final quality
enacarbil and gabapentin to be similarly effective against based on new evidence and trials published before our
postherpetic neuralgia [29], and one placebo-controlled trial of search date). The recommendation for the use of pre-
gabapentin ER gave negative results for failed back surgery gabalin is weak, as most recent high-quality trials have
syndrome with predominant neuropathic pain [30]. These recent yielded negative results, with a balance between efficacy
studies suggest that gabapentin is effective but are inconclusive and adverse effects lower than that for gabapentin, based
for gabapentin ER. The final quality of evidence was high. on a high-quality comparative trial. The evidence for the
We identified 25 placebo-controlled trials of pregabalin (18 use of other antiepileptics is inconclusive, although
yielding positive results) published before June 2013, with a oxcarbazepine and lacosamide may be beneficial for
high final quality of evidence. We identified 14 more recent small selected subgroups of patients (low-quality of evi-
high-to-very high-quality RCTs for pregabalin (150–600 mg/ dence). Based on our inclusion criteria, we found no data
day; 2101 patients treated) in the context of peripheral concerning clonazepam [39] in neuropathic pain. This
neuropathic pain, mostly due to diabetic polyneuropathy, does not rule out a potential efficacy of clonazepam on
including 12 placebo-controlled trials in which pregabalin pain paroxysms as reported in open-label uncontrolled
efficacy was primary outcome or pregabalin was an active studies [40] and might have a clinical interest in case of
comparator (secondary outcome) (Appendix 5). Only two of the noctural awakening by the pain.
eight trials considering pregabalin efficacy as the primary
outcome yielded positive results (137 patients treated). Two
revue neurologique 176 (2020) 325–352 331

3.2.3. Opioids negative results for the primary outcome, but included several
We identified 13 placebo-controlled trials of strong opioids confounding factors (Appendix 5). Two of the three studies
(mostly morphine and oxycodone, with 10 trials yielding concluded that lidocaine plasters had a positive effect in
positive results) and seven placebo-controlled trials of patients with peripheral localized neuropathic pain, particu-
tramadol (all yielding positive results) published before June larly those with allodynia, with a moderate final quality of
2013, with a moderate final quality of evidence. We also evidence.
identified two controlled trials of the opioid agonist and We identified seven placebo-controlled studies of high-
norepinephrine reuptake inhibitor tapentadol (one yielding concentration capsaicin patches (five yielding positive
weakly positive results), with a low-quality of evidence. We results) and 11 studies of low-concentration capsaicin cream
also found six additional placebo-controlled or comparative (seven yielding positive results) for peripheral neuropathic
studies of opioids (transdermal buprenorphine, fentanyl pain published before June 2013, with a high final quality of
patch and tapentadol) in 683 patients, one of which assessed evidence for high-concentration capsaicin patches and a
neuropathic pain in a heterogeneous group of patients with low-quality of evidence for low-concentration capsaicin
pain. One of these studies was a high-quality placebo- patches. We identified four more recent RCTs (351 patients
controlled study of transdermal buprenorphine in diabetic treated) of topical capsaicin in peripheral neuropathic pain:
painful neuropathy, which yielded negative results for the two moderate-quality non-inferiority trials reported similar
primary outcome [in the intention-to-treat (ITT) population], effects for low-concentration (0.025% to 0.075%) capsaicin
but positive results for most secondary outcomes, including cream and for topical clonidine or amitriptyline [49,50]; one
efficacy in the per protocol population [41]. One moderate- moderate-quality semi-double-blind placebo-controlled
quality placebo-controlled study of fentanyl one-day patches study showed that low-concentration (0.625%) capsaicin
for chronic pain yielded positive results for the primary patches were superior to placebo, whereas high-concentra-
outcome, but negative results in patients with a neuropathic tion (1.25%) patches were not [51]; one high-quality study of
pain component [42,43]. One study on tapentadol with an high-concentration capsaicin (8%) patches yielded positive
enriched enrollment design yielded positive results for results, albeit with a modest effect size (7% absolute
diabetic painful neuropathy, with a small effect size (NNT difference in daily pain scores) in diabetic painful neuropa-
9.1) [44], and a moderate-quality double-blind comparative thy [52]. In all these studies, the placebo patches were
study reported similar efficacies for 500 mg/day tapentadol identical in appearance to the patches delivering active
monotherapy and a combination of tapentadol (300 mg/day) treatment, but it was not possible to guarantee patient
and pregabalin (300 mg/day) in patients with low back pain blinding because skin reactions frequently occur with
with a neuropathic component [45]. The results of these capsaicin. These recent data suggest that high-concentra-
recent studies were inconclusive for buprenorphine and tion capsaicin patches are effective against diabetic peri-
fentanyl, and favored the use of tapentadol with a moderate pheral neuropathic pain, with a moderate final quality of
final quality of evidence. No recent RCT of tramadol was evidence, whereas the results for low-concentration cap-
identified. saicin cream or patches are inconclusive.
Three moderate-quality studies [53–55] and one low-
quality study [56] yielded positive results for herbal prepara-
tions or nitroglycerin. One high-quality study on topical
Recommendation: Strong opioids (particularly morphine ketamine yielded negative results for multiple-etiology peri-
and oxycodone) and tramadol have a weak recommen- pheral neuropathic pain [57]. The final quality of evidence was
dation for use in neuropathic pain (based on studies low.
published before our search date). The evidence for
buprenorphine and fentanyl is inconclusive (moderate-
quality trials). Tapentadol has a weak recommendation
for use in peripheral neuropathic pain (moderate final Recommendation: High-concentration capsaicin pat-
quality of evidence). ches have a weak recommendation for use in peripheral
neuropathic pain, including diabetic neuropathic pain
(high-quality of evidence, but low effect size). The rec-
ommendation for the use of topical lidocaine in periph-
3.2.4. Topical agents eral neuropathic pain is weak (moderate quality of
We identified three placebo-controlled studies of 5% lidocaine evidence but excellent safety profile). The evidence for
plasters (one yielding positive results) published before June the use of capsaicin at low concentrations, herbal pre-
2013, with a low final quality of evidence. Three more recent parations, nitroglycerin, clonidine and topical ketamine
high-quality placebo-controlled RCTs assessed lidocaine is inconclusive.
plasters (250 patients treated) in patients with postsurgical
or multiple-etiology neuropathic pain [46–48]: two of these
studies [46,48] yielded positive results. Pickering et al. reported
positive effects of lidocaine patches on mechanical and cold 3.2.5. Botulinum toxin A
allodynia after nerve surgery (the primary outcome in this We identified five low- to moderate-quality placebo-control-
study). The largest (multicenter, 180 treated patients and 183 led trials of subcutaneous (SC) botulinum toxin A in peripheral
controls) study [47] on postsurgical neuropathic pain gave neuropathic pain published before June 2013 (all five yielding
332 revue neurologique 176 (2020) 325–352

positive results, plus one large unpublished trial yielding a moderate final quality of evidence against their use. We
negative results). Three more recent placebo-controlled identified two additional more recent high-quality RCTs of
studies assessed the use of SC botulinum toxin A in peripheral cannabinoids in neuropathic pain: one study of nabiximols
neuropathic pain or spinal cord injury pain (74 patients (Sativex, 128 patients treated) yielded positive results for
treated). One of these studies was of low-quality and the other peripheral neuropathic pain, albeit with a modest effect size
two were of high- to very high-quality, and all yielded positive (NNT = 8.3); another study of oral dronabinol (tetrahydrocan-
results [58,59]. The highest quality study found that the nabinol or THC, 124 patients treated) yielded negative results
presence and severity of allodynia and residual intraepider- for neuropathic pain due to multiple sclerosis [62,63]. The final
mal nerve fiber density on skin punch biopsy predicted the quality of evidence was high.
response to BTX-A and that this drug was also particularly
effective against mechanical allodynia and paroxysmal pain
[59]. The other high-quality RCT showed that BTX-A was
effective against below-level spinal cord injury pain [60]. These Recommendation: The evidence concerning the level of
two recent studies support the efficacy of BTX-A against recommendation for the use of cannabinoids (transmu-
neuropathic pain, with a high final quality of evidence. cosal nabiximols, oral THC) in neuropathic pain is incon-
clusive due to large number of high-quality trials yielding
negative results.

Recommendation: Despite two recent positive high-


quality trials, the recommendation for SC botulinum
toxin A in peripheral neuropathic pain remains weak, 3.2.8. New drugs
due to the paucity of high-quality large-scale studies. One moderate-quality RCT compared cebranopadol, a
novel drug combining nociceptin/orphanin peptide and
opioid receptor agonism, to tapentadol and placebo in
chronic low back pain with a neuropathic component, and
3.2.6. Combination therapy reported similar efficacies in patients with and without
We identified 13 RCTs of combination therapy in neuropathic neuropathic pain components [43]. One study of the
pain (10 positive) published before June 2013, including three alpha2delta ligand mirogabalin yielded positive results
high-quality studies highlighting the relevance of combining for diabetic painful neuropathy [64]. One study reported a
pregabalin with duloxetine, or gabapentin with opioids or significant effect of the NGF antagonist fulranumab in
TCAs for the treatment of neuropathic pain. We identified diabetic painful neuropathy, whereas another reported
three more recent double-blind RCTs of combination therapy, negative results for this drug in post-traumatic neuropa-
two of which included TCAs (108 patients). One high-quality thic pain [65,66], but these studies were stopped pre-
study found that the combination of nortriptyline with maturely due to potential safety concerns with this drug
morphine at moderate doses was more effective than class (rapidly progressive osteoarthritis). One study of a
monotherapy at higher doses for the treatment of peripheral topical Nav1.7 antagonist in postherpetic neuralgia yielded
neuropathic pain [23,24]. Both these studies concluded that negative results, but this drug was found to be beneficial
the combination of TCAs with morphine or pregabalin was in 63% of patients carrying a particular Nav1.7 polymor-
more effective than monotherapy, with a high final quality of phism versus only 35% of patients without this variant
evidence. One small double-blind active comparative study (14 [67]. One study of an orally administered Nav1.7 antago-
patients per group) reported similar efficacies for methadone, nist in painful diabetic polyneuropathy reported negative
oral ketamine and their combination in refractory neuropathic results overall, but positive results for patients with
pain [61]. ‘‘burning pain’’ based on the NPSI [68]. Two phase II
studies of an angiotensin type II antagonist were stopped
prematurely in April 2019 due to safety issues in monkeys.

Recommendation: There is a weak recommendation for 3.2.9. Lack of data


the combination of antidepressants (SNRIs or TCAs) with Based on our inclusion criteria, we were unable to obtain data
gabapentinoids or morphine and for the combination of for the use of nefopam [69], paracetamol [70], NSAIDs [71] and
gabapentinoids and opioids in neuropathic pain, based midazolam in neuropathic pain.
on evidence dating from before 2013 and the results of
two more recent high-quality trials. The evidence for 3.3. Oral ketamine, intravenous drugs, nerve blocks and
other combinations is inconclusive. intrathecal therapy

3.3.1. Oral ketamine


We identified two double-blind studies of oral ketamine
3.2.7. Cannabinoids published since 1966 (135 patients treated): one high-quality
We identified nine placebo-controlled studies on cannabi- placebo-controlled study in patients with cancer neuropathic
noids (mostly transmucosal nabiximols and oral THC, only pain, which reported negative results for the primary outcome
two yielding positive results) published before June 2013, with [72], and one moderate-quality comparative study of oral
revue neurologique 176 (2020) 325–352 333

methadone, ketamine and their combination in neuropathic


pain refractory to conventional therapy, which reported a
significant improvement in pain relative to baseline of similar
magnitude in all three groups [61]. However, in this second Recommendation: The evidence was inconclusive
study, only ketamine decreased allodynia and burning pain. concerning the level of recommendation for nerve blocks
The final quality of evidence was low. in neuropathic pain.

Recommendation: The evidence is inconclusive for the 3.3.4. Intrathecal therapy


use of oral ketamine in neuropathic pain. We identified two double-blind placebo-controlled studies of
intrathecal (IT) methylprednisolone for neuropathic pain
performed on patients with intractable postherpetic neuralgia
(139 patients treated) since 1966. The first was initially
3.3.2. Intravenous drugs considered to be of high-quality [80], but was subsequently
We identified three placebo-controlled studies of intravenous downgraded to moderate quality as the results were incons-
(IV) lidocaine (70 patients treated) published since 1966. Two of istent with those of other studies of PHN and clinical
these trials used continuous lidocaine infusions (from 3 to experience. This study has since been heavily criticized [14].
7.5 mg/kg) once weekly for four consecutive weeks, one in An independent group was unable to reproduce its results and
diabetic neuropathic pain (low-quality trial) [73] and the other preclinical data are against IT corticosteroid efficacy [81,82]. In
in a mixed group (postherpetic neuralgia and CRPS; high- the second study, IT methylprednisolone (60 mg), midazolam
quality trial) [74]. These trials yielded positive results at the (2 mg) and their combination were assessed in postherpetic
end of the fourth week, but this effect did not persist four neuralgia involving lumbosacral dermatomes. The combina-
weeks later. However, the highest quality trial (very high) [75] tion was superior to each of the drugs delivered as a
evaluating the efficacy of a single infusion (5 mg/kg) gave monotherapy [83]. The final quality of evidence was low to
negative results at week 4. moderate.
We identified only one placebo-controlled study assessing
the long-term efficacy of IV ketamine in neuropathic pain
published since 1966. In this single-center moderate-quality
study of 40 patients randomized to two groups, a five-hour Recommendation: The evidence is inconclusive regard-
daily infusion of ketamine (80 mg/day) every day for one week ing the level of recommendation for intrathecal therapy
as an adjuvant to gabapentin was effective against spinal cord with methylprednisolone for neuropathic pain. We
injury pain one and two weeks after the last infusion, but found no data for ziconotide, morphine and clonidine
negative results were obtained at three weeks [76]. in neuropathic pain, as no double-blind study specifically
Overall, studies of iv lidocaine and ketamine generally assessing neuropathic pain has been published. This
supported short-term efficacy for these drugs (i.e. one to three does not rule out potential efficacy for IT morphine
weeks after the infusion) but a lack of long-term efficacy and ziconotide, as these drugs have been shown to be
(beyond three to four weeks), with a low-to-moderate final beneficial in double-blind trials in patients with refrac-
quality of evidence. No data were found for other iv drugs, tory chronic pain, including patients with neuropathic
including, in particular B12 injections [77]. pain [84,85].

Recommendation: The evidence concerning the level of 3.4. Neurostimulation therapy and invasive procedures
recommendation for iv lidocaine and ketamine in neu-
ropathic pain is inconclusive. However, given the poten- 3.4.1. Peripheral noninvasive neurostimulation
tial short-term efficacy of these drugs after their infusion, We identified nine RCTs of transcutaneous electrical nerve
they might be useful for dealing with acute exacerbations stimulation (TENS) published since 1966: one of high-quality,
of neuropathic pain (low-quality of evidence). six of moderate quality and two of low-quality (332 actively
treated patients) [86–94]. All but one of these studies assessed
peripheral neuropathic pain (the exception concerned spinal
cord injury pain) and were sham-controlled (six studies) or
3.3.3. Nerve blocks had an active control group (three studies). One study had a
We identified two high-quality studies fulfilling our inclusion double-blind design and seven used a single-blind design
criteria published since 1966 (including 289 patients). Both (three with patient blinding and four with investigator
used a combination of lidocaine and methylprednisolone, but blinding). Eight studies, including the double-blind study,
they gave opposite results [78,79]. One gave negative results gave positive results for pain, albeit with a modest effect size.
for pudendal neuralgia, whereas the other gave positive One other low-quality RCT evaluated the impact of local
results for the treatment of pain due to various nerve injuries. injections of cobalamine, lidocaine or both in addition to
The final quality of evidence was moderate. TENS. TENS seemed to be more effective when combined with
334 revue neurologique 176 (2020) 325–352

local injections of cobalamine [95]. Tolerability was excellent, neuropathic pain due to multiple sclerosis gave positive
with only minor skin irritations reported in some patients. results [114].
These studies were generally in favor of TENS as an effective Cranial electrotherapy stimulation (CES) involves the
treatment for peripheral neuropathic pain, with a moderate application of a small amount of current through the head
quality of evidence. via ear-clip electrodes. Two studies conducted by the same
We identified one study of static magnetic field treatment group investigated the effect of CES in a total of 67 stimulated
for painful diabetic neuropathy, which yielded positive results patients with spinal cord injury [115,116]. Both yielded positive
for the primary outcome [96]. Two well-conducted double- results for pain, but with a small effect size. The final quality of
blind placebo-controlled studies of pulsed magnetic field evidence was low.
treatments in the same population gave negative results
[97,98]. Two double-blind studies conducted by the same
group investigated the impact of frequency-modulated elec-
tromagnetic neural stimulation (FREMS) for painful diabetic Recommendation: There is a weak recommendation for
neuropathy [99,100]. These FREMS studies included a total of the use of high-frequency rTMS of the motor cortex in
85 actively treated patients, and the largest study yielded neuropathic pain (moderate final quality of evidence).
negative results. These studies generally supported a lack of The evidence concerning the level of recommendation of
efficacy of pulsed magnetic field and FREMS treatments, with a rTMS for DLPFC, tDCS, tsDCS and CES is inconclusive
low final quality of evidence. (low-quality of evidence), and there is a weak recommen-
dation against the use of rTMS of the posterior insula and
anterior cingulate cortex in neuropathic pain (one high-
quality study with negative results).
Recommendation: There is a weak recommendation for
the use of TENS in peripheral neuropathic pain (moderate
final quality of evidence). There is a weak recommenda-
tion against the use of static or pulsed magnetic fields 3.4.3. Pulsed radiofrequency (PRF)
(low final quality). PRF works by delivering a targeted electrical field to the
dorsal root ganglion (DRG) via a catheter needle tip, without
causing damage, as the temperature is maintained at or
below 42 8C [117]. By contrast, conventional radiofrequency
3.4.2. Central noninvasive neurostimulation thermocoagulation exposes the target nerves to continuous
We identified 16 placebo-controlled studies of central non- electrical stimulation and produces a lesion by increasing
invasive neurostimulation in various neuropathic pain condi- the temperature around the tip of the needle. We identified
tions. Seven studies of low-to-moderate quality used 12 double-blind studies of PRF for radicular pain or post-
repetitive transcranial magnetic stimulation (rTMS) of the herpetic neuralgia (PHN) in 383 actively treated patients
primary motor cortex (M1) on a total of 121 actively stimulated [118–129]. Six of these studies were of high-quality, with four
patients [101–107]. Protocols were heterogeneous in terms of yielding positive results [119,121,123,129]. One of these
the stimulation frequency used (5 to 20 Hz), number of pulses studies with positive results concerned cervical radicular
per session (500 to 2000), and number of sessions (5 or 10). One pain, one concerned lumbosacral radicular pain and the
study involving a small number of pulses (500) showed pain other two related to thoracic PHN. The two high-quality
reduction only immediately after stimulation [103], whereas studies yielding negative results for pain concerned lumbo-
all the others reported prolonged analgesia, for at least 15 days sacral radicular pain [118,122]. The six lower quality studies
after the end of sessions, and for at least seven weeks in four either gave positive results for pain or reported non-
studies [101,102,105,107]. These studies generally supported inferiority relative to the comparative group (conventional
the efficacy of high-frequency rTMS of the motor cortex for the radiofrequency in one case and percutaneous nucleoplasty
treatment of neuropathic pain, with a moderate final quality of in the other). No major adverse effects were reported. These
evidence. One small study in which the novel Hesed coil studies were in favor of pulsed radiofrequency as an
(‘‘deep rTMS’’) device was used in 25 patients yielded positive effective treatment for thoracic PHN, with a moderate
results [108]. Two studies targeting the left dorsolateral quality of evidence.
prefrontal cortex (DLPF) (n = 21) [109] or the posterior insula
and anterior cingulate cortex (n = 98) [110] gave negative
results (moderate final quality of evidence).
Three low- to moderate-quality studies used transcranial Recommendation: There is a weak recommendation for
direct current stimulation (tDCS) with anodal M1 montage in a the use of pulsed radiofrequency in thoracic postherpetic
total of 46 actively stimulated patients [111–113]. One study neuralgia (moderate final quality of evidence). The data
showed analgesia for four weeks after five daily sessions, are inconclusive for radicular pain (two studies yielding
whereas the other two yielded negative results. These studies positive results and two yielding negative results, all of
were generally supported a lack of efficacy of tDCS, but the high-quality).
final quality of evidence was low. One small, low-quality study
of transcutaneous spinal direct current stimulation (tsDCS) in
revue neurologique 176 (2020) 325–352 335

3.4.4. Peripheral invasive nerve stimulation study, which included 198 patients and was performed much
We identified two sham-controlled single-blind studies of more recently [137]. In this last study, the most frequent
percutaneous electrical nerve stimulation (PENS) applied for adverse event was implant site pain, which affected 11% of
three consecutive weeks (30 minutes, 3 times a week), to treat patients, and no SCS-related serious adverse events were
peripheral neuropathic pain due to sciatica or diabetic reported. The risk of spinal cord damage, through direct
neuropathy, yielding positive results for pain intensity lesions or indirect compression by epidural hematoma, is
[88,130]. In one study, PENS was also significantly more exceptionally low. The final quality of evidence was
effective than TENS for improving sleep and function. The moderate.
quality of evidence was considered low, as both studies were Based on our inclusion criteria (including at least three
single-blind and performed by the same group. One low- weeks of follow-up in a controlled clinical trial), we identified
quality study of implanted peripheral nerve stimulation in 45 only one RCT of invasive epidural motor cortex stimulation
actively treated patients yielded positive results [131]. One (EMCS) with double-blind assessment over one month,
moderate-quality open-label trial compared SCS and dorsal involving a comparison of the ON and OFF conditions, in a
root ganglion (DRG) stimulation [132]. A subgroup analysis of total of 13 patients [140]. The pain rating index of the McGill
patients with neuropathic pain and peripheral nerve lesions Pain Questionnaire (secondary endpoint) was significantly
suggested that both techniques were effective, with DRG lower in the ON than in the OFF condition, but no effect on pain
stimulation at least non-inferior to SCS. intensity (primary endpoint) was observed. The quality of
evidence was low. By contrast, previous uncontrolled open-
label studies had suggested that EMCS (frequency 20–65 Hz,
amplitude 1–6 V, pulse width 60–450 ms) might be beneficial,
Recommendation: The evidence concerning the level of particularly in patients undergoing several sessions of high-
recommendation for peripheral invasive nerve stimula- frequency rTMS of the motor cortex.
tion, including DRG stimulation for the treatment of We identified no studies meeting our inclusion criteria and
neuropathic pain is inconclusive. providing data concerning the efficacy of deep brain stimula-
tion in neuropathic pain.

3.4.5. Central invasive neurostimulation


We identified seven moderate-quality open-label controlled Recommendation: All the controlled studies of conven-
studies [133–138] and one high-quality double-blind RCT tional SCS in failed back surgery syndrome and diabetic
[139] of spinal cord stimulation (SCS) using conventional or neuropathic pain yielded positive results, with large
recently developed stimulation paradigms for failed back effect sizes and a low risk of adverse effects, but these
surgery syndrome or diabetic painful neuropathy. Four studies were open-label and generally compared SCS to
studies of conventional SCS for failed back surgery syndrome standard care. This resulted in a weak GRADE recom-
or diabetic painful neuropathy, involving a total of 144 mendation for the use of conventional SCS in neuropath-
patients with SCS [133–136], gave positive results with a large ic pain associated with failed back surgery syndrome and
effect size (NNT of 1.7 to 2.8): these studies compared SCS to diabetic painful neuropathies (moderate final quality of
repeated lumbosacral spine surgery in one study [133] and evidence). The evidence concerning the level of recom-
conventional medical management in the other three mendation for invasive motor cortex stimulation in neu-
studies. Two subsequent open-label RCTs involving 297 ropathic pain is inconclusive (low-quality of evidence).
patients stimulated with recently developed paradigms
(paresthesia-free, high-frequency stimulation at 10 kHz)
reported high levels of efficacy relative to conventional
parameters (usually around 80 Hz) for failed back surgery
3.4.6. Other surgical interventions
syndromes or back and leg pain [137,138]. One moderate-
quality open-label trial compared SCS and dorsal root We identified four controlled open-label studies of various
ganglion (DRG) stimulation [132]. A subgroup analysis of surgical interventions for neuropathic pain [141–144]. There
patients with neuropathic pain and peripheral nerve lesions was only one high-quality study, which concerned percuta-
suggested that both techniques were effective, with DRG neous epidural adhesiolysis in chronic lumbar radicular pain
stimulation at least non-inferior to SCS. One small double- and yielded positive results [142]. The other three studies
blind study of 24 patients with failed back surgery syndrome were of low-to-moderate quality and evaluated long-wave
found a significant effect of very high-frequency stimulation diathermy and interferential currents in chemotherapy-
(5882 Hz) versus sham stimulation, whereas stimulation at induced neuropathy (negative), nerve decompression in
lower frequencies (1200 Hz and 3030 Hz) was no more painful diabetic neuropathy (positive), and decompression
effective than sham stimulation [139]. The most frequent and transposition of the pudendal nerve in pudendal
complications were related to hardware dysfunction (e.g. neuralgia (positive). The final quality of evidence was low
migration, disconnection, electrode breakage) potentially to moderate.
requiring additional surgery for up to 24% of patients in one No eligible study concerning the microsurgical approach to
study conducted 15 years ago [134], but only 4% in the largest the dorsal root entry zone (DREZotomy) was identified.
336 revue neurologique 176 (2020) 325–352

of vitamin E supplementation in patients with diabetic painful


Recommendation: The evidence was inconclusive neuropathy reported negative results [155], as did a high-quality
concerning the level of recommendation for epidural study in the same population [156]. One low-quality study
adhesiolysis, long-wave diathermy and nerve decom- reported a small non-significant benefit of exercise versus
pression in neuropathic pain. There were no data education for peripheral neuropathic pain [157].
concerning DREZotomy. This does not rule out its poten- Based on our inclusion criteria, we identified no eligible
tial efficacy in spinal nerve root pain (brachial plexus studies concerning the use of mirror therapy, musicotherapy,
avulsion or cauda equina injury), as reported in uncon- art therapy, hypnotherapy, physiotherapy, mesotherapy,
trolled open-label studies [145,146]. homeopathy, therapeutic patient education, tai chi or Chinese
acupuncture.

3.5. Psychotherapy
Recommendation: The evidence concerning the level of
We identified five open-label RCTs of psychotherapy for recommendation for aromatherapy, auricular acupunc-
neuropathic pain: three concerned cognitive behavioral the- ture and low-level laser therapy was inconclusive, as it
rapy (CBT) in a total of 89 patients [147–149] and two related to was obtained in single low- or moderate-quality trials,
mindfulness in 46 patients [150,151]. One study used supportive and there is a weak recommendation against the use of
psychotherapy as a control group [147], whereas the other four vitamin E in neuropathic pain. There were no data
used usual care alone (waiting list). Two low-quality CBT concerning other complementary therapies. This does
studies on group therapy for neuropathic pain related to HIV not rule out the potential efficacy of mirror therapy,
[147] or spinal cord injury [148] reported positive effects on pain hypnosis and acupuncture in neuropathic pain, which
intensity. The third moderate-quality study [149] of a web- have been evaluated in randomized controlled studies in
based CBT intervention (10 modules, including videos, exercise which neuropathic pain was assessed at early time
sheets to be downloaded, navigation on the dedicated website, points (before 3 weeks) or in trials not focusing on neu-
available only to study participants) reported a decrease in pain ropathic pain [158–160].
intensity in chemotherapy-induced painful neuropathies. This
study also suggested that web-based interventions are feasible
in this context. Two RCTs assessed the efficacy of mindfulness
versus usual care in patients with neuropathic pain. The first of 3.7. Studies in pediatric or elderly populations
these studies was a low-quality pilot study of individual daily
meditation sessions for six weeks; it reported negative results Only one small comparative study was identified in children
for pain intensity in elderly patients with postherpetic [161]. It showed that both 10 mg/day amitriptyline and 900 mg/
neuralgia [150]. By contrast, the second study was a high- day gabapentin decreased pain relative to baseline. There was
quality study of group therapy as part of a mindfulness-based no significant difference between treatments and no major side
stress reduction program (MBSR) in patients with painful effects. We identified only one specific study in elderly patients
diabetic neuropathy; it demonstrated clear efficacy, with a (mean age: 72 years) focusing on mindfulness (see above).
large effect (NNT = 2.4) [151]. Both studies reported an effect of
mindfulness on quality of life, function and catastrophizing. 3.8. Therapeutic algorithm in adults
The final quality of evidence was moderate.
Details regarding the GRADE recommendations are provided
in Tables 1–3 and Fig. 2. Table 4 presents practical recom-
mendation for use. All the proposals were deemed appropriate
Recommendation: There is a weak recommendation for by the reading group (median  7). Relative (range: 5 to 9) or
the use of cognitive behavior therapy and mindfulness as strong (range: 7 to 9) agreement of at least 90% of reading group
an add-on therapy for neuropathic pain (moderate final members [162] was obtained for all recommendations except
quality of evidence). the one for carbamazepine (Appendix 6).

4. Discussion
3.6. Other miscellaneous non-drug treatments
This manuscript presents French recommendations for the
Seven studies using various nondrug treatments were identi- treatment of neuropathic pain based on a comprehensive
fied. One low-quality study compared aromatherapy massage systematic review of pharmacological and non-pharmacolo-
to usual care alone and suggested a small improvement of pain gical therapies. Given the heterogeneous nature of the
and quality of life [152]. One moderate-quality study of auricular treatments and designs considered here, we did not conduct
acupuncture for SCI pain reported positive results [153]. One a new meta-analysis and do not, therefore, report the effect
low-quality study investigated the impact of low-level laser sizes or number needed to treat (NNT) for a group of therapies.
treatment for postherpetic neuralgia and reported positive However, we have provided these quantitative data, when
results relative to the placebo [154]. One moderate-quality study available, for individual studies (see Appendix 5). We used the
revue neurologique 176 (2020) 325–352 337

Table 1 – Guidelines for pharmacotherapy (available in France) based on grade.


Grade Drugs Daily dosages/regimenRecommendations Specific safety issues (warning)
Strong for Duloxetine 60–120 mg/day First line Contraindicated with monoamine
oxidase inhibitors, and in cases of hepatic
or severe renal failure. Suicidal behavior
(low risk), due to disinhibiting effects
Venlafaxine 150–225 mg/day First line
Gabapentin 1200–3600 mg/day First line Potential abuse
TCAs 25–150 mg/day First line (2nd line in the elderly) Contraindicated with monoamine
oxidase inhibitors, and in cases of closed-
angle glaucoma risk, urinary retention
risk or recent myocardial infarction.
Suicidal behavior (low risk) due to
disinhibiting effect
Weak for Lidocaine plasters 1–3 plasters for 12 h/day First line for peripheral NP Contraindicated on broken skin
Pregabalin 150–600 mg/day Second line (alternative to Potential abuse
gabapentin)
Capsaicin 8% 1–4 patches/3 months Second line for peripheral NP Available in a limited number of pain
units
BTX-A (SC) 50–300 U/3 months Second line for NP Available in a limited number of pain
units
Combination TCA (or SNRIs) + opioids Second or third line (if opioids) Greater risk for combinations of
or + gabapentinoids gabapentinoids with opioids (even
tramadol)
Tramadol 100–400 mg/day Second line Potential abuse. Contraindicated with
monoamine oxidase inhibitors, and in
cases of severe respiratory failure
Strong opioids < 150 MME/day Third line (in absence of Potential abuse. Greater risk when
alternative) combined with gabapentinoids
Inconclusive Capsaicin cream
Carbamazepine
Clonidine topical
Lacosamidea
Oxcarbazepineb
IV ketamine
IV lidocainec
Lamotrigine
SSRIs
Topiramate
Zonisamide
Cannabinoids
Weak against Valproate
Strong against Levetiracetam
Mexiletine
Lack of data for Nefopam paracetamol
neuropathic NSAIDs clonazepam
pain Midazolam
Vitamin B12
Only treatments available in France are included in this table.
What does GRADE recommendation mean?: Strong for: high-quality evidence that the desirable effects of the intervention are clearly greater
than its undesirable effects. Weak for or weak against: uncertainty about the trade-offs (because of low- to moderate-quality evidence or
because the desirable and undesirable effects balance out). Generally inconclusive: not enough data to conclude for neuropathic pain in
general. This does not mean that these treatments are or are not effective, and they can be proposed in specific situations. Strong against:
high-quality evidence that the desirable effects are clearly not greater than the undesirable effects. Lack of data: absence of studies
corresponding to the inclusion criteria. This does not mean that the treatments are effective or ineffective.
TCAs: tricyclic antidepressants; BTX: botulinum toxin; MME: morphine milligram equivalent; TCA: tricyclic antidepressant; SSRIs: selective
serotonin reuptake inhibitors; BID: bis in die (= twice daily); NSAIDs: nonsteroidal anti-inflammatory drugs; GRADE: grading of
recommendations assessment, development and evaluation.
a
Could be of interest in a specific population of patients with Nav1.7 mutation-related small-fiber neuropathy (based on a single study).
b
Could be of interest in patients with peripheral neuropathy and ‘‘irritable nociceptor’’ phenotype (i.e., limited thermal deficit) (based on a
single study).
c
IV ketamine (0.25 to 1 mg/kg per day) and IV lidocaine (3–7.5 mg/kg, once weekly) may be proposed to overcome acute pain exacerbations in
patients with neuropathic pain refractory to more conventional medication, because of their short-lasting analgesic effects (less than 4 weeks
in double-blind trials using these doses and modes of administration). Careful cardiac monitoring is warranted with lidocaine. Misuse and
long-term side effects (particularly renal and hepatic toxicity) should be monitored in cases of repeated ketamine infusions.
338 revue neurologique 176 (2020) 325–352

Table 2 – Intrathecal therapy, neurostimulation and surgery.


Grade Treatment Regimen Recommendations Specific safety issues
(warning)
Weak for TENS  30 min/day First line Seizures (HF-rTMS)
HF-rTMS over M1 > 1200 pulses/session, > 5 Hz Third line
Pulsed radiofrequency Paresthesia elicited along the Thoracic post-herpetic
tested nerve root at low intensities; neuralgia
PRF settings 42 8C, 120 seconds/
twice for the same level
Spinal cord stimulation Conventional (40 to 80 Hz) or high- Third line (FBSS or diabetic
frequency stimulation (without polyneuropathy)
paresthesia, 1 to 10 kHz)
Inconclusive Intrathecal
methylprednisolone
Anodal M1 tDCS
CES
PENS
EMCS
HF-rTMS over DLPFC
Weak against HF-rTMS over posterior
insula or ACC
Pulsed magnetic field
FREMS
Lack of data for Intrathecal morphine,
neuropathic pain ziconotide or clonidine
DBS
TENS: transcutaneous electrical nerve stimulation; HF-rTMS: high-frequency repetitive transcranial magnetic stimulation; M1: primary motor
cortex; IT: intrathecal; IV: intravenous; FBSS: failed back surgery syndrome; tDCS: transcutaneous direct current stimulation; PENS:
percutaneous electrical nerve stimulation; EMCS: epidural motor cortex stimulation; DBS: deep brain stimulation; DLPFC: dorsolateral
prefrontal cortex; ACC: anterior cingulate cortex; FREMS: frequency-modulated electromagnetic neural stimulation; CES: cranial electro-
therapy stimulation.
What does GRADE recommendation mean?: Strong for: high-quality evidence that the desirable effects of the intervention are clearly greater
than its undesirable effects. Weak for or weak against: uncertainty about the trade-offs (because of low- to moderate-quality evidence or
because the desirable and undesirable effects balance out). Generally inconclusive: not enough data to conclude for neuropathic pain in
general. This does not mean that these treatments are or are not effective, and they can be proposed in specific situations. Strong against:
high-quality evidence that the desirable effects are clearly not greater than the undesirable effects. Lack of data: absence of studies
corresponding to the inclusion criteria. This does not mean that the treatments are effective or ineffective.

Table 3 – Psychotherapies and complementary therapies.


Grade Treatment Regimen Recommendations Specific safety issues (warning)
Weak for Cognitive behavioral therapy As an adjuvant treatment
Mindfulness-based stress reduction
Weak against Vitamin E
Lack of data for Other psychotherapies
neuropathic pain Massage
Aromatherapy
Auriculotherapy
Musicotherapy
Art therapy
Hypnotherapya
Physiotherapy
Mesotherapy
Mirror therapya
Homeopathy
Tai chi
Chinese acupuncturea
What does GRADE recommendation mean?: Strong for: high-quality evidence that the desirable effects of the intervention are clearly greater
than its undesirable effects. Weak for or weak against: uncertainty about the trade-offs (because of low- to moderate-quality evidence or
because the desirable and undesirable effects balance out). Generally inconclusive: not enough data to conclude for neuropathic pain in
general. This does not mean that these treatments are or are not effective, and they can be proposed in specific situations. Strong against:
high-quality evidence that the desirable effects are clearly not greater than the undesirable effects. Lack of data: absence of studies
corresponding to the inclusion criteria. This does not mean that the treatments are effective or ineffective.
a
See Table 4 for additional information regarding these treatments.
revue neurologique 176 (2020) 325–352 339

Fig. 2 – Proposed therapeutic algorithm for neuropathic pain treatment in adults. Only treatments available in France in June
2019 were considered. For practical considerations, see Table 4. NP: neuropathic pain; TCAs: tricyclic antidepressants;
TENS: transcutaneous electrical nerve stimulation; HF-rTMS: high-frequency repetitive transcranial magnetic stimulation;
M1: primary motor cortex; BTX-A: botulinum toxin type A; FBSS: failed back surgery syndrome; MME: mg morphine
equivalent.

GRADE system to assess the quality of evidence for all pregabalin were proposed as first-line treatments, duloxetine
treatments, and our recommendations comply with the and lidocaine patches as second-line treatments and strong
AGREE II guidelines [20,163]. The GRADE system makes it opioids as third-line treatments. The most recent meta-
possible to combine studies of heterogeneous quality, because analysis and systematic study of pharmacotherapy establis-
it allows the upgrading of controlled open-label studies with a hed the efficacy of TCAs, SNRIs, gabapentin, pregabalin,
high final quality and the downgrading of double-blind studies tramadol and strong opioids for neuropathic pain, and that of
with many biases [163]. topical agents (topical lidocaine, high-concentration capsaicin
The major strength of this systematic review is that it is the and botulinum toxin A) for peripheral neuropathic pain [13].
first based on a comprehensive assessment of all therapies This analysis provided strong GRADE recommendations for
proposed for neuropathic pain treatment, encompassing the use of antidepressants, pregabalin and gabapentin, which
pharmacological treatments and all other types of other were proposed as first-line treatments, and a weak GRADE
medical/surgical treatment. All previous international recom- recommendation for topical agents and opioids, which were
mendations for neuropathic pain focused on a specific proposed as second-line (for topical agents and tramadol) or
treatment, such as pharmacotherapy [13], neurostimulation third-line (for strong opioids and botulinum toxin A) treat-
[15] or invasive therapy [14]. We also propose evidence-based ments, but they did not recommend combination therapy
recommendations for psychotherapies and complementary because few high-quality studies were available at the time
medicine. [13]. Since the completion of this previous systematic study,
Our therapeutic algorithm for pharmacotherapy is driven we have identified 22 studies of monotherapy with TCAs,
by the drug treatment rather than by the etiology or duloxetine, gabapentin, lidocaine plasters or botulinum toxin
peripheral/central lesion responsible for pain, as in the 2015 A providing positive results for neuropathic pain and which
NeuPSIG recommendations [13] and previous French recom- were generally of high-quality, and one moderate-quality
mendations [12]. Indeed, neuropathic pain is now fully study of high-concentration capsaicin patches that also
recognized to have multiple etiologies across similar neuropa- yielded positive results. No new studies of venlafaxine were
thic syndromes [164,165]. In previous French recommenda- identified. Hence, the level of recommendation based on
tions [12] based on a systematic review, TCAs, gabapentin and GRADE remains strong for tricyclic antidepressants (at
340 revue neurologique 176 (2020) 325–352

Table 4 – Practical use of treatments for chronic neuropathic pain available in France.
First line for focal peripheral First-line treatments for focal peripheral neuropathic pain include: lidocaine plasters, 1
neuropathic pain to 3 plasters depending on the extent of pain, 12 hours per day. Adverse effects mostly
skin irritation or allergy; ttranscutaneous electrical pain stimulation. Reimbursed if
prescribed by specialist pain units in France. Adverse effects mostly skin irritation or
allergy
These topical therapies may be used as standalone treatments or in combination with
systemic drugs (expert consensus) and should be preferred in patients with limited area
of pain and residual sensitivity (i.e. lack of total anesthesia to thermal or mechanical
stimulation of the painful area)
First line for any neuropathic pain First-line treatments for any neuropathic pain include: oral SNRIs: duloxetine (60–
120 mg per day once daily or BID) or venlafaxine (150–225 mg daily administered BID or
TID) with a preference for duloxetine (based on higher quality trials with positive
results). Main adverse effects include nausea, decreased appetite, constipation and
sexual side effects. Rare cases of high blood pressure have been reported with high
doses of venlafaxine ( 150 mg daily). Suicidal ideation is rare, but possible on initiation
of any antidepressants (warning); oral gabapentin (1200–3600 mg administered TID).
Main adverse effects include drowsiness, dizziness, weight gain and peripheral edema.
Misuse risk factors should be investigated before prescription of gabapentin and
potential misuse or abuse should be assessed at each renewal because of the potential
for the misuse of this drug; oral TCAs (i.e., amitriptyline, clomipramine, imipramine),
10–150 mg per day administered once daily or BID. Low doses (< 75 mg/d) are usually
effective. Common adverse effects include drowsiness and weight gain. Suicidal
ideation is rare, but possible at initiation of any antidepressant (warning). TCAs should
be carefully monitored for cardiac effects, anticholinergic effects or postural
hypotension, especially in polymedicated patients, patients with multiple diseases and
at doses > 75 mg daily. In the elderly, polymedicated adults or adults with multiple
diseases, doses should not exceed 75 mg per day
The ‘‘start low and go slow’’ approach is essential for all orally administered drugs, to
minimize adverse effects and patients should be advised of the risk of an impairment of
their ability to drive, particularly at treatment initiation
Second line for focal peripheral Second-line treatments for focal peripheral neuropathic pain include: capsaicin, high-
neuropathic pain concentration patches (1 to 4 patches depending on the extent of pain, applied for
30 minutes on the feet or 60 minutes for other body areas). Main adverse effects include
acute burning pain after application (which may be reduced by cold applications),
redness and swelling. Rare cases of high blood pressure have been reported because of
the acute pain induced by application. Patients should therefore be monitored for acute
pain and blood pressure for at least one hour after application; botulinum toxin A (up to
300 units subcutaneously into the painful area, depending on the extent of the pain).
Adverse effects consist principally of pain due to the injection
These drugs are available for hospital use only (and at the day hospital for capsaicin).
They may be used as standalone treatments or combined with systemic drugs (expert
consensus) and should be preferred in patients with a limited area of pain, residual
sensitivity (i.e. lack of total anesthesia to thermal or mechanical stimulation of the
painful area), and mechanical allodynia for botulinum toxin A. Botulinum toxin A can
be prescribed only by specialists
Second line for any neuropathic Second-line treatments for any neuropathic pain include: oral pregabalin (150–600 mg
pain daily, administered BID or TID as an alternative to gabapentin in cases of poor
tolerability or inefficacy). Main adverse effects include drowsiness, dizziness, weight
gain and peripheral edema. Misuse risk factors should be investigated before
prescription and potential misuse or abuse should be assessed at each renewal because
of the potential for abuse (apparently more commonly than with gabapentin);
sustained-release oral tramadol (100–400 mg daily BID or once daily; a single once daily
preparation is available for use). Tramadol has the same side effects and potential abuse
risks as strong opioids (see below), although such risks are less common, as the
maximum dose of tramadol (400 mg/day), corresponds to 40 to 68 mg morphine
equivalent (evidence.nhs.uk). Tramadol may be combined with antiepileptics (expert
consensus), but should be used with caution with high-dose antidepressants (potential
risk of serotoninergic syndrome); combination of antidepressants (TCAs 25–75 mg daily
or duloxetine 60 mg daily) with gabapentinoids (pregabalin 150–300 mg daily or
gabapentin 1200–1800 mg daily); cognitive behavioral therapy and mindfulness. These
techniques have the highest level of evidence amongst psychotherapies and are
recommended as add-ons to other therapies (as controlled studies showing their
efficacy were conducted in patients receiving pharmacotherapy)
The ‘‘start low and go slow’’ approach is essential for all orally administered drugs, to
minimize adverse effects, and patients should be advised of the risk of an impairment
of their ability to drive, particularly at treatment initiation
revue neurologique 176 (2020) 325–352 341

Third line Third-line treatments for neuropathic pain include: high-frequency rTMS of the motor
cortex (80% of the motor threshold, 10 Hz, 1500 to 3000 pulses per session) can be
proposed for localized or more diffuse pain (i.e. spinal cord injury or diabetic painful
neuropathy). This technique is available only at specialized centers. The principal
adverse effects are headache within hours of the session. Seizures are exceptional
(warning); spinal cord stimulation (SCS) has been found to be effective in failed back
surgery syndrome and painful diabetic polyneuropathy. It is available only at
specialized centers. It may be considered before initiating strong opioids and after as
little as one year of pain. Like other invasive techniques, SCS should always be preceded
by careful psychological assessment of the patient. Complications are rare and are
mostly related to hardware dysfunction (e.g. migration, disconnection, electrode
breakage) potentially requiring additional surgery. The risk of spinal cord damage, due
to a direct lesion or indirect compression by epidural hematoma, is exceptionally low;
strong opioids, i.e. sustained-release oral morphine and oxycodone. Strong opioids
should be proposed in the absence of alternatives. They require careful evaluation of
potential risks of abuse, preferably with specific questionnaires, such as the opioid risk
tool, and should be used for the shortest possible duration (i.e. a few months), with
careful monitoring, and, preferably, with initiation by pain specialists. Their doses
should be titrated up slowly and should not exceed 150 mg morphine equivalent (expert
consensus). Immediate-release opioids should be avoided, particularly transmucosal
fentanyl (except in cases of associated cancer based on official approval for use) because
of the higher risk of abuse associated with these preparations. In the lack of substantial
improvements in pain, function or quality of life, opioids should be titrated down and
stopped. The main adverse effects include constipation, drowsiness, nausea, pruritus,
dysuria. Long-term opioid use is also associated with misuse, abuse, hypogonadism and
hypocortisolism [213]. Misuse risk factors should be investigated before prescription
and misuse or abuse should be assessed at each renewal. Opioid prescription should be
monitored particularly carefully in men under the age of 50 years, due to the endocrine
and misuse risks; combination of opioids with antidepressants or gabapentinoids, in
the absence of alternatives. Opioid doses (including tramadol) should be reduced in
combination with gabapentinoids due to recent reports of a greater risk of death with
such combinations [169]
Practical recommendations for No study has defined the rules for switching to second- or third-line therapy in
switch in treatment neuropathic pain. In the absence of contraindication, first-line treatments should be
tested first. In cases of intolerable adverse effects or treatment failure, defined as a
decrease in pain of less than 30% [214] and/or an improvement in function of less than
30% after a trial for at least six weeks at the maximum tolerated dose, a switch to
second-line therapy should be proposed (expert consensus). The same rules should
apply when switching from second- to third-line therapies (expert consensus)
TENS: transcutaneous electrical nerve stimulation; TCAs: tricyclic antidepressants; BID: bis in die (= twice daily); TID: ter in die (three times
daily); BTX-A: botulinum toxin type A; rTMS: repetitive transcranial magnetic stimulation; SCS: spinal cord stimulation; EMCS: epidural motor
cortex stimulation.

relevance of combining gabapentin with morphine for the


moderate doses), gabapentin and SNRIs, which we recom- treatment of neuropathic pain, we recommend this combina-
mend as first-line treatments, and weak for capsaicin patches, tion as a third-line treatment, in the absence of suitable
which we recommend as a second-line treatment for peri- alternatives, with the proviso that it should be carefully
pheral neuropathic pain and localized pain (Fig. 2). Based on monitored, with prescription of the lowest possible doses, as
new high-quality studies reporting positive results for BTX-A suggested by a well-conducted epidemiological retrospective
and on a good efficacy/safety ratio for both treatments study reporting a higher risk of opioid-related death in patients
[166,167], we increased the GRADE level of recommendation taking such a combination [169]. Low dose combination can be
for topical lidocaine and botulinum toxin A, which we now effective and better tolerated than high dose monotherapy. No
recommend as first- and second-line treatments, respectively, more recent studies on tramadol were identified. The level of
for peripheral neuropathic pain in which the pain is confined recommendation for tramadol therefore remains unchanged
to a limited area. Based on recent high-quality combination relative to the previous guidelines [13].
studies showing that combinations of TCAs with opioids or Contrary to all previous recommendations, we now
with gabapentinoids are superior to monotherapy with these propose pregabalin as an alternative to gabapentin for
agents, we now also recommend combinations of anti- second-line treatment. Indeed, most recent large-scale high-
depressants and gabapentinoids as a second-line treatment quality studies have shown pregabalin to be ineffective or only
if insufficient pain relief is achieved with monotherapy. This weakly effective relative to placebo at doses similar to those
recommendation extends to the combination of SNRI anti- used in routine clinical practice (generally 300 mg daily). All
depressants with pregabalin, based on one large-scale high- these studies, including those in which positive results were
quality trial giving positive results before the start date for our obtained, had very small effect sizes. A recent high-quality
search [168]. Again, based on one high-quality study dating comparative study in chronic sciatica showed that gabapentin
from before the start of our search and demonstrating the was significantly more effective and had fewer adverse effects
342 revue neurologique 176 (2020) 325–352

than pregabalin [36]. This is also consistent with recent meta- treatments considered inconclusive or ‘‘weak against’’ in prior
analyses reporting a lower number needed to harm (NNH) for meta-analyses generally remained similar in this analysis, in
pregabalin (13.9) than for gabapentin (25.6) [13]. Another high- the absence of new evidence for efficacy or safety.
quality study reported that pregabalin lacked efficacy against We also considered clinical trials performed specifically in
acute and chronic sciatica [170]. Finally, the COMBO trial children or in elderly or frail patients with neuropathic pain.
comparing the efficacy of monotherapy with pregabalin or However, we were unable to find any high-quality studies
duloxetine to the combination of these two drugs in diabetic complying with our criteria. Our guidelines for these popu-
painful neuropathy also reported a significantly lower efficacy lations are, therefore, mostly based on expert consensus.
for pregabalin (300 mg daily) than for duloxetine (60 mg daily) Thus, we recommend gabapentin (10 to 30 mg/kg per day, TID)
in the first phase of the trial [168]. Overall, these data suggest or amitriptyline (0.3 to 1 mg/kg/day once per day) as the first-
that the efficacy and safety of pregabalin was overestimated in line treatment for neuropathic pain in children. Other drug
initial studies. Furthermore, recent evidence has revealed a treatments recommended for adults may also be proposed for
potential for abuse for drugs of this class [171–173]. Abuse is this population, including topical lidocaine, in particular, as a
increasingly reported for gabapentinoids and abuse rates first-line treatment for peripheral neuropathic pain in which
seem to be higher for pregabalin than for gabapentin [174–176]. the pain is localized to a particular area. In cases of insufficient
We therefore recommend a careful monitoring of gabapenti- pain relief, a combination of gabapentin and amitriptyline or a
noids. Prescribers should also be aware of high-risk popula- treatment switch is recommended. Psychological support and
tions (i.e. younger patients, patients with a history of physiotherapy to improve function and physical development
substance abuse, particularly opioids, and psychiatric comor- should always be proposed either on their own or as add-on
bidities) and should monitor for signs of abuse, as for opioids treatments. In cases of first-line treatment failure, the patient
[177]. Of note, in the UK, gabapentinoids have been reclassified may be referred to a pediatric chronic pain clinic. All other
as a class C-controlled drug since April 2019.</span></span> drug treatments or invasive procedures should be proposed
Our recommendations regarding opioids should be seen in only after interdisciplinary consultation. Further studies are
the context of the dramatic opioid crisis in the USA, where required to improve the quality of evidence for this popula-
misuse rates and opioid-related mortality are high [178,179]. tion, despite the difficulty of performing large-scale studies in
Indeed, prescription opioids were responsible for more than this context [182–184]. The possible contribution of multi-
17,000 deaths in the USA in 2017 (drugabuse.gov). The disciplinary pain treatment programs should also be consi-
situation is much less severe in France, but opioid prescription dered.
rates are increasing [178,180], and guidelines should be In the elderly, slow titration is particularly important for
followed to limit the risk of misuse [177]. The evidence for systemic treatment [185,186]. In this population, a consensus-
opioid efficacy and safety in neuropathic pain is stronger than based algorithm for neuropathic pain management has been
for other chronic pain syndromes, such as osteoarthritis and proposed, with gabapentinoids, lidocaine plasters and dulo-
back pain [177]. However, recent studies of strong opioids in xetine as first-line treatments [185]. Given the lack of specific
neuropathic pain were limited to fentanyl and transdermal trials in the elderly, we propose to use this algorithm for first-
buprenorphine, and their results were inconclusive, contrast- line therapy, with a switch to TCAs as the second-line
ing with the positive results obtained for oral morphine and treatment, due to specific safety concerns (cardiac arrythmias
oxycodone in previous meta-analyses and systematic reviews and anticholinergic effects) in this population [187].
[12,13,177]. We therefore recommend oral sustained-release As reported in previous meta-analyses [13,188], most
morphine or oxycodone as a third-line treatment for neuropa- pharmacological treatments, even those proposed as first-
thic pain, in the absence of suitable alternatives, with specific line treatments, are of limited efficacy, with an effect size < 0.5
warnings and careful monitoring (expert consensus). Opioids [189] or a NNT > 7. There may be many reasons for this,
should be prescribed for the shortest possible duration and including drugs not necessarily been used in the right
withdrawal anticipated (see Table 4 for practical use). population [4]. For this reason, an increasing number of
Tapentadol, a newer agent combining opioid agonism and clinical trials in neuropathic pain are stratifying patients on
norepinephrine reuptake properties, is probably effective the basis of sensory phenotype or taking phenotypes into
against peripheral neuropathic pain, according to the results account as secondary outcome measures for improving
of recent studies, but is not yet available in France. treatment outcome (e.g. Demant et al., 2014, 2015; Attal
Cannabinoids are under investigation for use against et al., 2016). In particular, lacosamide seems to be relevant for
refractory neuropathic pain in France at specialized pain use in patients with Nav1.7 mutation-related small-fiber
centers [181]. The previous recommendations [13] were neuropathy, based on the findings of a single study [38], and
‘‘weakly against’’ their use, because of potential risks of abuse oxcarbazepine has been reported to be effective in patients
and the larger number of trials giving negative than positive with an ‘‘irritable nociceptor’’ sensory phenotype [37]. We
results. Based on two recent trials, one reporting weakly strongly encourage similar studies in the future, with the aim
positive findings for nabiximols and with the other negative of targeting specific drug therapies on the basis of phenotypes
results for dronabinol, the evidence was considered incon- and underlying mechanisms.
clusive, because there are still more studies giving negative Unlike previous evidence-based systematic reviews, this
than positive results, for both compounds. This does not review analyzed all potential drug treatments for neuropathic
exclude the possibility that cannabinoids act in subgroups of pain, including those used as single administrations, such as
patients or on specific symptoms, particularly paroxysmal IV ketamine or lidocaine. These drugs are widely used at pain
pain. The level of recommendation regarding other drug centers in France, for refractory chronic neuropathic pain, in
revue neurologique 176 (2020) 325–352 343

the absence of alternatives, despite considerable heteroge- spinal cord stimulation for failed back surgery syndrome and
neity in terms of dose and administration route for ketamine diabetic neuropathic pain. Although not evaluated in the
(‘‘OKAPI’’, Pickering, NCT03319238). The evidence for keta- studies included in this analysis, there is also a potential
mine was considered inconclusive due to the very small interest in the use of this technique for other painful
number of studies (only one) meeting our criteria, which polyneuropathies. The development of new SCS techniques
included, in particular, pain assessment at three weeks. using, for example, very high-frequency stimulation, which
However, this does not rule out its potential benefit at earlier can generate analgesia without paresthesia, should improve
time points, for up to two weeks after infusion, as reported in the level of evidence through sham-controlled double-blind
previous studies (e.g. Amr, 2010). Moreover, ketamine may studies, such as those beginning to emerge [139,199,200].
also be beneficial for major depression and suicidal ideation, These sham-controlled studies highlight the importance of
which may be associated with severe pain [190]. We found no the placebo effect associated with such procedures.
evidence for the efficacy of single injections of IV lidocaine, but Epidural motor cortex stimulation (MCS) is another
two studies using repeated administrations gave positive invasive technique, for which a double-blind procedure is
results, albeit with no long-lasting effects. Furthermore, as for feasible. Previous meta-analyses of MCS using less stringent
ketamine, positive results were often obtained at early time criteria concluded that the procedure was safe and probably
points (1–2 weeks) in these studies, and in previous trials [191– effective against neuropathic pain [15,201–203]. This syste-
194]. Based on these findings and expert consensus, we matic review identified only one sham-controlled double-
propose the use of IV ketamine or lidocaine to overcome acute blind study complying with our inclusion criteria: this study
pain exacerbations in chronic neuropathic pain refractory to had equivocal results (negative for the primary outcome and
more conventional medication. We strongly encourage fur- positive for the secondary outcomes) [140]. Two other low-
ther well-designed studies, including long-term assessments, quality double-blind studies with very small sample sizes
for these drug treatments. yielded positive results but did not satisfy our inclusion
Our systematic review also encompassed all stimulation criteria due to their short follow-up period [204] or retro-
techniques, some of which, such as TENS, are widely proposed spective design [205]. Therefore, in the absence of recent high-
for neuropathic pain as a standalone therapy or in addition to quality studies, and based on the potential, limited risks
pharmacotherapy. Based on the GRADE system (recommen- associated with this technique, the evidence for motor cortex
dation ‘‘weak for’’), we recommend TENS as a first-line stimulation was rated as inconclusive in this recommenda-
treatment for peripheral NP (only one studied yielded positive tion. Furthermore, the availability of this technique is
results for central pain). The quality of evidence for non- restricted to a few trained surgical teams. However, open-
invasive brain neurostimulation (rTMS) was limited by the label studies have suggested that the outcome of the invasive
lack of long-term sham-controlled assessments of analgesic procedure might be improved by selecting candidates for
effects in most cases. Nonetheless, one sham-controlled study implantation according to their preoperative analgesic res-
based on a six-month protocol reported sustained efficacy for ponse to several sessions of noninvasive high-frequency rTMS
motor cortex rTMS in a series of 20 patients with fibromyalgia of the motor cortex [206–209]. The use of such procedures and
[195], and real-life data are also consistent with long-term the development of new designs for stimulation or electrodes
beneficial effects of motor cortex rTMS in various types of [210] are awaited, to establish the relevance of invasive motor
neuropathic pain, for maintenance sessions [196–198]. We cortex stimulation in the routine management of neuropathic
therefore propose a weak recommendation for high-fre- pain.
quency rTMS of the motor cortex in refractory neuropathic Our recommendations also took into account psychothe-
pain. rapies, complementary approaches, such as hypnosis or
Invasive therapies for neuropathic pain include pulsed acupuncture, and therapeutic education of the patient. These
radiofrequency, intrathecal drugs, spinal cord stimulation and therapies are generally included in multidisciplinary inter-
epidural motor cortex stimulation. The efficacy of radio- ventions, and only a very small number of the programs
frequency has been confirmed by high-quality studies in a widely used in clinical practice for neuropathic pain [e.g.
selected population of patients with thoracic postherpetic psychodynamic psychotherapy, hypnotherapy, eye move-
neuralgia. As such, it can be recommended as a third-line ment desensitization and reprocessing (EMDR), mindfulness]
treatment in this very specific population. Strikingly, although met our inclusion criteria. Nonetheless, based on their
the level of evidence for intrathecal therapy is moderate to potential benefit in other chronic pain disorders [211–214]
high in chronic pain in general, very few data were available and on the basis of the few available studies meeting our
concerning the efficacy of this therapy specifically against requirements, we recommend CBT and mindfulness as
neuropathic pain, and the level of evidence was therefore second-line treatments in addition to other therapies, because
considered inconclusive [84]. of their safety, and high levels of acceptability and feasibility,
Long-term prospective controlled studies of spinal cord even in children or elderly patients.
stimulation were conducted several years ago and the
procedure is generally considered safe, but the quality of
these studies remains moderate at best, because of the 5. Conclusion
impossibility of blinding, due to the paresthesia induced by
the stimulation. Indeed, the control group in the highest The goal of these comprehensive recommendations was to
quality study published to date had no intervention [134]. For rationalize the use of all therapeutic options by general
these reasons, the level of recommendation remains weak for practitioners and physicians involved in pain management in
344 revue neurologique 176 (2020) 325–352

France, and on a broader scale. The dissemination of these


guidelines will make a major contribution in this direction. Appendix A. Supplementary data
Indeed, a large study of general practitioners’ adherence to the
2010 French recommendations revealed poor recall of appro- Supplementary data associated with this article can be
priate first-line drugs three years after the publication of these found, in the online version, at https://doi.org/10.1016/j.
guidelines [9]. We obtained similar findings for pain specialists neurol.2020.01.361.
[7]. We hope that these recommendations will help to improve
this outcome in the next few years.

references
Disclosure of interest

[1] Doth AH, Hansson PT, Jensen MP, Taylor RS. The burden of
X. Moisset has received personal fees from Teva, Novartis,
neuropathic pain: a systematic review and meta-analysis
Roche, Biogen, Sanofi-Genzyme and Merck-Serono, and non-
of health utilities. Pain 2010;149:338–44. http://dx.doi.org/
financial support from SOS oxygene, Boehringer, Bristol Myers 10.1016/j.pain.2010.02.034.
Squibb, not related to the submitted work. [2] Attal N, Lanteri-Minet M, Laurent B, Fermanian J,
H. Alchaar has received honoraria from Novartis, Sanofi, Bouhassira D. The specific disease burden of neuropathic
MSD, outside the submitted work in the past 5 years. pain: results of a French nationwide survey. Pain
J. Avez Couturier, S. Conradi and C. Regis declare that they 2011;152:2836–43. http://dx.doi.org/10.1016/
j.pain.2011.09.014.
have no competing interest.
[3] Langley PC, Van Litsenburg C, Cappelleri JC, Carroll D. The
D. Bouhassira has received honoraria from Grunenthal, burden associated with neuropathic pain in Western
Novartis and Air liquide, outside the submitted work and is Europe. J Med Econ 2013;16:85–95. http://dx.doi.org/
participating to European projects Dolorisk and IMI PainCare. 10.3111/13696998.2012.729548.
M.H. Delmotte has received honoraria from Mylan, outside [4] Colloca L, Ludman T, Bouhassira D, Baron R, Dickenson
the submitted work. AH, Yarnitsky D, et al. Neuropathic pain. Nat Rev Dis
Primer 2017;3:17002. http://dx.doi.org/10.1038/nrdp.2017.2.
M. Lanteri-Minet received grants and honoraria for
[5] Dworkin RH, Malone DC, Panarites CJ, Armstrong EP,
advisory boards, speaker panels or investigation studies from
Pham SV. Impact of postherpetic neuralgia and painful
Allergan, Amgen, Astellas, ATI, BMS, Boehringer, Boston diabetic peripheral neuropathy on health care costs. J Pain
Scientific, CoLucid, Convergence, GlaxoSmithKline, Grunen- 2010;11:360–8. http://dx.doi.org/10.1016/
thal, Eli Lilly, Medtronic, Menarini, MSD, Novartis, Pfizer, j.jpain.2009.08.005.
Reckitt Benckiser, Saint-Jude, Sanofi-Aventis, Teva Pharma- [6] Bouhassira D, Letanoux M, Hartemann A. Chronic pain
ceuticals, UCB, and Zambon. with neuropathic characteristics in diabetic patients: a
French cross-sectional study. PloS One 2013;8:e74195.
J.P. Lefaucheur has received honoraria from Novo Nordisk,
http://dx.doi.org/10.1371/journal.pone.0074195.
Pfizer, and Sanofi in the last 5 years, not related to the
[7] Piano V, Lanteri-Minet M, Steegers M, Besse K, Donnet A,
submitted work. Verhagen S, et al. A case vignette study to assess the
G. Mick has received honoraria from Sanofi MSD, Lilly, knowledge of pain physicians of neuropathic cancer pain:
Pfizer, Teva, Grunenthal, Mylan, BMS UPSA outside the room for improvement. Pain Physician 2013;16:E779–88.
submitted work in the past 5 years. [8] Torrance N, Ferguson JA, Afolabi E, Bennett MI, Serpell MG,
V. Piano has received honoraria from Novartis, Grünenthal, Dunn KM, et al. Neuropathic pain in the community: more
under-treated than refractory? Pain 2013;154:690–9. http://
Ethipharm, Mylan, not related to the submitted work.
dx.doi.org/10.1016/j.pain.2012.12.022.
G. Pickering has received honoraria from Sanofi, MSD, [9] Martinez V, Attal N, Vanzo B, Vicaut E, Gautier JM,
Grunenthal, Mylan, Kyowa-Kirin in the last 5 years, not related Bouhassira D, et al. Adherence of French GPs to chronic
to the submitted work. neuropathic pain clinical guidelines: results of a cross-
E. Piquet received honoraria from Grunenthal and Med- sectional, randomized, ‘‘e’’ case-vignette survey. PloS One
tronic, not related to the submitted work. 2014;9:e93855. http://dx.doi.org/10.1371/
journal.pone.0093855.
E. Salvat has received fees from Novartis, Sanofi, Ethy-
[10] Heutink M, Post MWM, Wollaars MM, van Asbeck FWA.
pharm, Grunenthal, CSL Behring, MSD, Merz, Kyowa-Kirin,
Chronic spinal cord injury pain: pharmacological and non-
Mundipharma, Mylan, Genzyme, Medtronic, Abbott and pharmacological treatments and treatment effectiveness.
Boston in the past 5 years, outside the submitted work. Disabil Rehabil 2011;33:433–40. http://dx.doi.org/10.3109/
N. Attal has received honoraria from Sanofi MSD, Sanofi- 09638288.2010.498557.
Aventis, Sanofi Pasteur MSD, MSD vaccins, Ethypharm, Lilly [11] Tamburin S, Lacerenza MR, Castelnuovo G, Agostini M,
France, Grunenthal, Ipsen, Astellas, outside the submitted Paolucci S, Bartolo M, et al. Pharmacological and non-
pharmacological strategies in the integrated treatment of
work, in the past 5 years and is a member of the European
pain in neurorehabilitation. Evidence and
DOLORISK consortium.
recommendations from the Italian Consensus Conference
on Pain in Neurorehabilitation. Eur J Phys Rehabil Med
2016;52:741–52.
Acknowledgments [12] Martinez V, Attal N, Bouhassira D, Lantéri-Minet M. Les
douleurs neuropathiques chroniques : diagnostic,
We would like to thank Nathalie Pinol-Domenech and évaluation et traitement en médecine ambulatoire.
Recommandations pour la pratique clinique de la Société
Manuela De Carvalho for their help with the literature search.
revue neurologique 176 (2020) 325–352 345

française d’étude et de traitement de la douleur. Douleurs [28] Enomoto H, Yasuda H, Nishiyori A, Fujikoshi S, Furukawa
Eval - Diagn - Trait 2010;11:3–21. http://dx.doi.org/10.1016/ M, Ishida M, et al. Duloxetine in patients with diabetic
j.douler.2009.12.009. peripheral neuropathic pain in Japan: a randomized,
[13] Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, double-blind, noninferiority comparative study with
Dworkin RH, et al. Pharmacotherapy for neuropathic pain pregabalin. J Pain Res 2018;11:1857–68. http://dx.doi.org/
in adults: a systematic review and meta-analysis. Lancet 10.2147/JPR.S170646.
Neurol 2015;14:162–73. http://dx.doi.org/10.1016/S1474- [29] Harden RN, Freeman R, Rainka M, Zhang L, Bell C, Berges
4422(14)70251-0. A, et al. A phase 2a, randomized, crossover trial of
[14] Dworkin RH, O’Connor AB, Kent J, Mackey SC, Raja SN, gabapentin enacarbil for the treatment of postherpetic
Stacey BR, et al. Interventional management of neuropathic neuralgia in gabapentin inadequate responders. Pain Med
pain: NeuPSIG recommendations. Pain 2013;154:2249–61. Malden Mass 2013;14:1918–32. http://dx.doi.org/10.1111/
http://dx.doi.org/10.1016/j.pain.2013.06.004. pme.12227.
[15] Cruccu G, Garcia-Larrea L, Hansson P, Keindl M, [30] Gewandter JS, Frazer ME, Cai X, Chiodo VF, Rast SA, Dugan
Lefaucheur J-P, Paulus W, et al. EAN guidelines on central M, et al. Extended-release gabapentin for failed back
neurostimulation therapy in chronic pain conditions. Eur J surgery syndrome: results from a randomized double-
Neurol 2016;23:1489–99. http://dx.doi.org/10.1111/ blind crossover study. Pain 2019;160(5):1029–36. http://
ene.13103. dx.doi.org/10.1097/j.pain.0000000000001478.
[16] Affsaps. Prise en charge médicamenteuse de la douleur [31] Mu Y, Liu X, Li Q, Chen K, Liu Y, Lv X, et al. Efficacy and
aiguë et chronique chez l’enfant; 2009. safety of pregabalin for painful diabetic peripheral
[17] Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, neuropathy in a population of Chinese patients: a
Alonso-Coello P, et al. GRADE: an emerging consensus on randomized placebo-controlled trial. J Diabetes
rating quality of evidence and strength of 2018;10:256–65. http://dx.doi.org/10.1111/1753-0407.12585.
recommendations. BMJ 2008;336:924–6. http://dx.doi.org/ [32] Markman J, Resnick M, Greenberg S, Katz N, Yang R,
10.1136/bmj.39489.470347.AD. Scavone J, et al. Efficacy of pregabalin in post-traumatic
[18] Balshem H, Helfand M, Schünemann HJ, Oxman AD, Kunz peripheral neuropathic pain: a randomized, double-blind,
R, Brozek J, et al. GRADE guidelines: 3. Rating the quality of placebo-controlled phase 3 trial. J Neurol 2018;265:2815–
evidence. J Clin Epidemiol 2011;64:401–6. http://dx.doi.org/ 24. http://dx.doi.org/10.1007/s00415-018-9063-9.
10.1016/j.jclinepi.2010.07.015. [33] Raskin P, Huffman C, Toth C, Asmus MJ, Messig M,
[19] Bendtsen L, Zakrzewska JM, Abbott J, Braschinsky M, Di Sanchez RJ, et al. Pregabalin in patients with inadequately
Stefano G, Donnet A, et al. EAN guideline on trigeminal treated painful diabetic peripheral neuropathy: a
neuralgia. Eur J Neurol 2019. http://dx.doi.org/10.1111/ randomized withdrawal trial. Clin J Pain 2014;30:379–90.
ene.13950. http://dx.doi.org/10.1097/AJP.0b013e31829ea1a1.
[20] Brouwers M, Kho M, Browman G. AGREE II: advancing [34] Huffman C, Stacey BR, Tuchman M, Burbridge C, Li C,
guideline development, reporting and evaluation in health Parsons B, et al. Efficacy and safety of pregabalin in the
care. CMAJ 2010;182:E839–42. treatment of patients with painful diabetic peripheral
[21] Haute Autorité de santé. Méthodes d’élaboration des neuropathy and pain on walking. Clin J Pain 2015;31:946–
recommandations de bonne pratique; 2014 [accessed May 58. http://dx.doi.org/10.1097/AJP.0000000000000198.
18, 2019]https://www.has-sante.fr/portail/jcms/c_418716/ [35] Raskin P, Huffman C, Yurkewicz L, Pauer L, Scavone JM,
fr/methodes-d-elaboration-des-recommandations-de- Yang R, et al. Pregabalin in patients with painful diabetic
bonne-pratique. peripheral neuropathy using an NSAID for other pain
[22] Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group. conditions: a double-blind crossover study. Clin J Pain
Preferred reporting items for systematic reviews and 2016;32:203–10. http://dx.doi.org/10.1097/
meta-analyses: the PRISMA statement. BMJ AJP.0000000000000254.
2009;339:b2535. [36] Robertson K, Marshman LAG, Plummer D, Downs E. Effect
[23] Gilron I, Tu D, Holden RR, Jackson AC, DuMerton-Shore D. of gabapentin vs pregabalin on pain intensity in adults
Combination of morphine with nortriptyline for with chronic sciatica: a randomized clinical trial. JAMA
neuropathic pain. Pain 2015;156:1440–8. http://dx.doi.org/ Neurol 2019;76:28–34. http://dx.doi.org/10.1001/
10.1097/j.pain.0000000000000149. jamaneurol.2018.3077.
[24] Holbech JV, Bach FW, Finnerup NB, Brøsen K, Jensen TS, [37] Demant DT, Lund K, Vollert J, Maier C, Segerdahl M,
Sindrup SH. Imipramine and pregabalin combination for Finnerup NB, et al. The effect of oxcarbazepine in
painful polyneuropathy: a randomized controlled trial. peripheral neuropathic pain depends on pain phenotype:
Pain 2015;156:958–66. http://dx.doi.org/10.1097/ a randomised, double-blind, placebo-controlled
j.pain.0000000000000143. phenotype-stratified study. Pain 2014;155:2263–73. http://
[25] Vollmer TL, Robinson MJ, Risser RC, Malcolm SK. A dx.doi.org/10.1016/j.pain.2014.08.014.
randomized, double-blind, placebo-controlled trial of [38] de Greef BTA, Hoeijmakers JGJ, Geerts M, Oakes M, Church
duloxetine for the treatment of pain in patients with TJE, Waxman SG, et al. Lacosamide in patients with Nav1.7
multiple sclerosis. Pain Pract 2014;14:732–44. http:// mutations-related small fibre neuropathy: a randomized
dx.doi.org/10.1111/papr.12127. controlled trial. Brain J Neurol 2019;142:263–75. http://
[26] Gao Y, Guo X, Han P, Li Q, Yang G, Qu S, et al. Treatment of dx.doi.org/10.1093/brain/awy329.
patients with diabetic peripheral neuropathic pain in [39] Corrigan R, Derry S, Wiffen PJ, Moore RA. Clonazepam for
China: a double-blind randomised trial of duloxetine vs. neuropathic pain and fibromyalgia in adults. Cochrane
placebo. Int J Clin Pract 2015;69:957–66. http://dx.doi.org/ Database Syst Rev 2012;CD009486. http://dx.doi.org/
10.1111/ijcp.12641. 10.1002/14651858.CD009486.pub2.
[27] Schukro RP, Oehmke MJ, Geroldinger A, Heinze G, Kress H- [40] Swerdlow M, Cundill JG. Anticonvulsant drugs used in the
G, Pramhas S. Efficacy of duloxetine in chronic low back treatment of lancinating pain. A comparison. Anaesthesia
pain with a neuropathic component: a randomized, 1981;36:1129–32. http://dx.doi.org/10.1111/j.1365-
double-blind, placebo-controlled crossover trial. 2044.1981.tb08701.x.
Anesthesiology 2016;124:150–8. http://dx.doi.org/10.1097/ [41] Simpson RW, Wlodarczyk JH. Transdermal buprenorphine
ALN.0000000000000902. relieves neuropathic pain: a randomized, double-blind,
346 revue neurologique 176 (2020) 325–352

parallel-group, placebo-controlled trial in diabetic neuropathy. J Diabetes Metab Disord 2015;14:86. http://
peripheral neuropathic pain. Diabetes Care 2016;39:1493– dx.doi.org/10.1186/s40200-015-0217-3.
500. http://dx.doi.org/10.2337/dc16-0123. [54] Heydari M, Homayouni K, Hashempur MH, Shams M.
[42] Arai T, Kashimoto Y, Ukyo Y, Tominaga Y, Imanaka K. Topical Citrullus colocynthis (bitter apple) extract oil in
Two placebo-controlled, randomized withdrawal studies painful diabetic neuropathy: a double-blind randomized
to evaluate the fentanyl 1 day patch in opioid-naı̈ve placebo-controlled clinical trial. J Diabetes 2016;8:246–52.
patients with chronic pain. Curr Med Res Opin http://dx.doi.org/10.1111/1753-0407.12287.
2015;31:2207–18. http://dx.doi.org/10.1185/ [55] Petramfar P, Moein M, Samani SM, Tabatabaei SH,
03007995.2015.1092127. Zarshenas MM. Trachyspermum ammi 10% topical cream
[43] Christoph A, Eerdekens M-H, Kok M, Volkers G, versus placebo on neuropathic pain, a randomized,
Freynhagen R. Cebranopadol, a novel first-in-class double-blind, placebo-controlled trial. Neurol Sci
analgesic drug candidate: first experience in patients with 2016;37:1449–55. http://dx.doi.org/10.1007/s10072-016-
chronic low back pain in a randomized clinical trial. Pain 2600-3.
2017;158:1813–24. http://dx.doi.org/10.1097/ [56] Viswanathan V, Rajsekar S, Selvaraj B, Kumpatla S. Effect
j.pain.0000000000000986. of a polyherbal formulation cream on diabetic
[44] Vinik AI, Shapiro DY, Rauschkolb C, Lange B, Karcher K, neuropathic pain among patients with type 2 diabetes – A
Pennett D, et al. A randomized withdrawal, placebo- pilot study. Indian J Med Res 2016;144:215–9. http://
controlled study evaluating the efficacy and tolerability of dx.doi.org/10.4103/0971-5916.195033.
tapentadol extended release in patients with chronic [57] Lynch ME, Clark AJ, Sawynok J, Sullivan MJL. Topical 2%
painful diabetic peripheral neuropathy. Diabetes Care amitriptyline and 1% ketamine in neuropathic pain
2014;37:2302–9. http://dx.doi.org/10.2337/dc13-2291. syndromes: a randomized, double-blind, placebo-
[45] Baron R, Martin-Mola E, Müller M, Dubois C, Falke D, controlled trial. Anesthesiology 2005;103:140–6.
Steigerwald I. Effectiveness and safety of tapentadol [58] Ghasemi M, Ansari M, Basiri K, Shaigannejad V. The
prolonged release (PR) versus a combination of tapentadol effects of intradermal botulinum toxin type a injections on
PR and pregabalin for the management of severe, chronic pain symptoms of patients with diabetic neuropathy. J Res
low back pain with a neuropathic component: a Med Sci 2014;19:106–11.
randomized, double-blind, phase 3b study. Pain Pract [59] Attal N, de Andrade DC, Adam F, Ranoux D, Teixeira MJ,
2015;15:455–70. http://dx.doi.org/10.1111/papr.12200. Galhardoni R, et al. Safety and efficacy of repeated
[46] Demant DT, Lund K, Finnerup NB, Vollert J, Maier C, injections of botulinum toxin A in peripheral neuropathic
Segerdahl MS, et al. Pain relief with lidocaine 5% patch in pain (BOTNEP): a randomised, double-blind, placebo-
localized peripheral neuropathic pain in relation to pain controlled trial. Lancet Neurol 2016;15:555–65. http://
phenotype: a randomised, double-blind, and placebo- dx.doi.org/10.1016/S1474-4422(16)00017-X.
controlled, phenotype panel study. Pain 2015;156:2234–44. [60] Han Z-A, Song DH, Oh H-M, Chung ME. Botulinum toxin
http://dx.doi.org/10.1097/j.pain.0000000000000266. type A for neuropathic pain in patients with spinal cord
[47] Palladini M, Boesl I, Koenig S, Buchheister B, Attal N. injury. Ann Neurol 2016;79:569–78. http://dx.doi.org/
Lidocaine-medicated plaster, an additional potential 10.1002/ana.24605.
treatment option for localized postsurgical neuropathic [61] Rigo FK, Trevisan G, Godoy MC, Rossato MF, Dalmolin GD,
pain: efficacy and safety results of a randomized, placebo- Silva MA, et al. Management of neuropathic chronic pain
controlled trial. Curr Med Res Opin 2019;35:757–66. http:// with methadone combined with ketamine: a randomized,
dx.doi.org/10.1080/03007995.2019.1565709. double-blind, active-controlled clinical trial. Pain
[48] Pickering G, Voute M, Macian N, Ganry H, Pereira B. Physician 2017;20:207–15.
Effectiveness and safety of 5% lidocaine-medicated plaster [62] Serpell M, Ratcliffe S, Hovorka J, Schofield M, Taylor L,
on localized neuropathic pain after knee surgery: a Lauder H, et al. A double-blind, randomized, placebo-
randomized, double-blind controlled trial. Pain controlled, parallel group study of THC/CBD spray in
2019;160:1186–95. http://dx.doi.org/10.1097/ peripheral neuropathic pain treatment. Eur J Pain
j.pain.0000000000001502. 2014;18:999–1012. http://dx.doi.org/10.1002/j.1532-
[49] Kiani J, Ahmad Nasrollahi S, Esna-Ashari F, Fallah P, Sajedi 2149.2013.00445.x.
F. Amitriptyline 2% cream vs. capsaicin 0.75% cream in the [63] Schimrigk S, Marziniak M, Neubauer C, Kugler EM, Werner
treatment of painful diabetic neuropathy (double-blind, G, Abramov-Sommariva D. Dronabinol is a safe long-term
randomized clinical trial of efficacy and safety). Iran J treatment option for neuropathic pain patients. Eur
Pharm Res IJPR 2015;14:1263–8. Neurol 2017;78:320–9. http://dx.doi.org/10.1159/000481089.
[50] Kiani J, Sajedi F, Nasrollahi SA, Esna-Ashari F. A [64] Vinik A, Rosenstock J, Sharma U, Feins K, Hsu C, Merante
randomized clinical trial of efficacy and safety of the D, et al. Efficacy and safety of mirogabalin (DS-5565) for
topical clonidine and capsaicin in the treatment of painful the treatment of diabetic peripheral neuropathic pain: a
diabetic neuropathy. J Res Med Sci 2015;20:359–63. randomized, double-blind, placebo- and active
[51] Moon J-Y, Lee P-B, Kim Y-C, Lee S-C, Nahm FS, Choi E. comparator-controlled, adaptive proof-of-concept phase 2
Efficacy and safety of 0.625% and 1.25% capsaicin patch in study. Diabetes Care 2014;37:3253–61. http://dx.doi.org/
peripheral neuropathic pain: multicenter, randomized, 10.2337/dc14-1044.
and semi-double-blind controlled study. Pain Physician [65] Wang H, Romano G, Frustaci ME, Bohidar N, Ma H, Sanga
2017;20:27–35. P, et al. Fulranumab for treatment of diabetic peripheral
[52] Simpson DM, Robinson-Papp J, Van J, Stoker M, Jacobs H, neuropathic pain: a randomized controlled trial.
Snijder RJ, et al. Capsaicin 8% patch in painful diabetic Neurology 2014;83:628–37. http://dx.doi.org/10.1212/
peripheral neuropathy: a randomized, double-blind, WNL.0000000000000686.
placebo-controlled study. J Pain 2017;18:42–53. http:// [66] Wang H, Romano G, Fedgchin M, Russell L, Sanga P, Kelly
dx.doi.org/10.1016/j.jpain.2016.09.008. KM, et al. Fulranumab in patients with pain associated
[53] Taheri A, Farbood A, Heshmat R, Samadi A, Khashayar P, with postherpetic neuralgia and postraumatic
Qorbani M, et al. The effect of transdermal nitroglycerin neuropathy: efficacy, safety, and tolerability results from a
on pain control in diabetic patients with peripheral randomized, double-blind, placebo-controlled, phase-2
revue neurologique 176 (2020) 325–352 347

study. Clin J Pain 2017;33:99–108. http://dx.doi.org/10.1097/ 2000;343:1514–9. http://dx.doi.org/10.1056/


AJP.0000000000000388. NEJM200011233432102.
[67] Price N, Namdari R, Neville J, Proctor KJW, Kaber S, Vest J, [81] Rijsdijk M, van Wijck AJM, Meulenhoff PCW, Kavelaars A,
et al. Safety and efficacy of a topical sodium channel van der Tweel I, Kalkman CJ. No beneficial effect of
inhibitor (TV-45070) in patients with postherpetic intrathecal methylprednisolone acetate in postherpetic
neuralgia (PHN): a randomized, controlled, proof-of- neuralgia patients. Eur J Pain Lond Engl 2013;17:714–23.
concept, crossover study, with a subgroup analysis of the http://dx.doi.org/10.1002/j.1532-2149.2012.00233.x.
Nav1.7 R1150W genotype. Clin J Pain 2017;33:310–8. http:// [82] Rijsdijk M, Svensson CI, Wijck AJvan, Kalkman CJ, Yaksh
dx.doi.org/10.1097/AJP.0000000000000408. TL. Analgesic properties of intrathecal glucocorticoids in
[68] McDonnell A, Collins S, Ali Z, Iavarone L, Surujbally R, three well established preclinical pain models. Scand J
Kirby S, et al. Efficacy of the Nav1.7 blocker PF-05089771 in Pain 2016;10:90–102. http://dx.doi.org/10.1016/
a randomised, placebo-controlled, double-blind clinical j.sjpain.2015.10.006.
study in subjects with painful diabetic peripheral [83] Dureja GP, Usmani H, Khan M, Tahseen M, Jamal A.
neuropathy. Pain 2018;159:1465–76. http://dx.doi.org/ Efficacy of intrathecal midazolam with or without
10.1097/j.pain.0000000000001227. epidural methylprednisolone for management of post-
[69] Joo YC, Ko ES, Cho JG, Ok YM, Jung GY, Kim KH. herpetic neuralgia involving lumbosacral dermatomes.
Intravenous nefopam reduces postherpetic neuralgia Pain Physician 2010;13:213–21.
during the titration of oral medications. Korean J Pain [84] Deer TR, Pope JE, Hayek SM, Bux A, Buchser E, Eldabe S,
2014;27:54–62. http://dx.doi.org/10.3344/ et al. The Polyanalgesic Consensus Conference (PACC):
kjp.2014.27.1.54. recommendations on intrathecal drug infusion systems
[70] Wiffen PJ, Knaggs R, Derry S, Cole P, Phillips T, Moore RA. best practices and guidelines. Neuromodulation J Int
Paracetamol (acetaminophen) with or without codeine or Neuromodulation Soc 2017;20:96–132. http://dx.doi.org/
dihydrocodeine for neuropathic pain in adults. Cochrane 10.1111/ner.12538.
Database Syst Rev 2016;12:CD012227. http://dx.doi.org/ [85] Deer TR, Pope JE, Hanes MC, McDowell GC. Intrathecal
10.1002/14651858.CD012227.pub2. therapy for chronic pain: a review of morphine and
[71] Moore RA, Chi C-C, Wiffen PJ, Derry S, Rice ASC. Oral ziconotide as first-line options. Pain Med Malden Mass
nonsteroidal anti-inflammatory drugs for neuropathic 2019;20:784–98. http://dx.doi.org/10.1093/pm/pny132.
pain. Cochrane Database Syst Rev 2015;CD010902. http:// [86] Rutgers M, van Romunde L, Osman P. A small randomized
dx.doi.org/10.1002/14651858.CD010902.pub2. comparative trial of acupuncture versus transcutaneous
[72] Fallon MT, Wilcock A, Kelly CA, Paul J, Lewsley L-A, Norrie electrical neurostimulation in postherpetic neuralgia. Pain
J, et al. Oral ketamine vs placebo in patients with cancer- Clin 1988;2:87–9.
related neuropathic pain: a randomized clinical trial. [87] Kumar D, Marshall HJ. Diabetic peripheral neuropathy:
JAMA Oncol 2018;4:870–2. http://dx.doi.org/10.1001/ amelioration of pain with transcutaneous
jamaoncol.2018.0131. electrostimulation. Diabetes Care 1997;20:1702–5.
[73] Viola V, Newnham HH, Simpson RW. Treatment of [88] Ghoname EA, White PF, Ahmed HE, Hamza MA, Craig WF,
intractable painful diabetic neuropathy with intravenous Noe CE. Percutaneous electrical nerve stimulation: an
lignocaine. J Diabetes Complications 2006;20:34–9. http:// alternative to TENS in the management of sciatica. Pain
dx.doi.org/10.1016/j.jdiacomp.2005.05.007. 1999;83:193–9.
[74] Kim Y-C, Castañeda AM, Lee C-S, Jin H-S, Park KS, Moon [89] Barbarisi M, Pace MC, Passavanti MB, Maisto M,
JY. Efficacy and safety of lidocaine infusion treatment for Mazzariello L, Pota V, et al. Pregabalin and transcutaneous
neuropathic pain: a randomized, double-blind, and electrical nerve stimulation for postherpetic neuralgia
placebo-controlled study. Reg Anesth Pain Med treatment. Clin J Pain 2010;26:567–72. http://dx.doi.org/
2018;43:415–24. http://dx.doi.org/10.1097/ 10.1097/AJP.0b013e3181dda1ac.
AAP.0000000000000741. [90] Gossrau G, Wähner M, Kuschke M, Konrad B, Reichmann
[75] Moulin DE, Morley-Forster PK, Pirani Z, Rohfritsch C, Stitt H, Wiedemann B, et al. Microcurrent transcutaneous
L. Intravenous lidocaine in the management of chronic electric nerve stimulation in painful diabetic neuropathy:
peripheral neuropathic pain: a randomized controlled a randomized placebo-controlled study. Pain Med Malden
trial. Can J Anaesth J Can Anesth 2019. http://dx.doi.org/ Mass 2011;12:953–60. http://dx.doi.org/10.1111/j.1526-
10.1007/s12630-019-01395-8. 4637.2011.01140.x.
[76] Amr YM. Multi-day low dose ketamine infusion as [91] Buchmuller A, Navez M, Milletre-Bernardin M, Pouplin S,
adjuvant to oral gabapentin in spinal cord injury related Presles E, Lantéri-Minet M, et al. Value of TENS for relief of
chronic pain: a prospective, randomized, double-blind chronic low back pain with or without radicular pain. Eur J
trial. Pain Physician 2010;13:245–9. Pain Lond Engl 2012;16:656–65. http://dx.doi.org/10.1002/
[77] Buesing S, Costa M, Schilling JM, Moeller-Bertram T. j.1532-2149.2011.00061.x.
Vitamin B12 as a treatment for pain. Pain Physician [92] Koca I, Boyaci A, Tutoglu A, Ucar M, Kocaturk O.
2019;22:E45–52. Assessment of the effectiveness of interferential current
[78] Eker HE, Cok OY, Aribogan A, Arslan G. Management of therapy and TENS in the management of carpal tunnel
neuropathic pain with methylprednisolone at the site of syndrome: a randomized controlled study. Rheumatol Int
nerve injury. Pain Med Malden Mass 2012;13:443–51. 2014;34:1639–45. http://dx.doi.org/10.1007/s00296-014-
http://dx.doi.org/10.1111/j.1526-4637.2011.01323.x. 3005-3.
[79] Labat JJ, Riant T, Lassaux A, Rioult B, Rabischong B, [93] Bi X, Lv H, Chen B-L, Li X, Wang X-Q. Effects of
Khalfallah M, et al. Adding corticosteroids to the pudendal transcutaneous electrical nerve stimulation on pain in
nerve block for pudendal neuralgia: a randomised, double- patients with spinal cord injury: a randomized controlled
blind, controlled trial. BJOG Int J Obstet Gynaecol trial. J Phys Ther Sci 2015;27:23–5. http://dx.doi.org/
2017;124:251–60. http://dx.doi.org/10.1111/1471- 10.1589/jpts.27.23.
0528.14222. [94] Naderi Nabi B, Sedighinejad A, Haghighi M, Biazar G,
[80] Kotani N, Kushikata T, Hashimoto H, Kimura F, Muraoka Hashemi M, Haddadi S, et al. Comparison of
M, Yodono M, et al. Intrathecal methylprednisolone for transcutaneous electrical nerve stimulation and pulsed
intractable postherpetic neuralgia. N Engl J Med radiofrequency sympathectomy for treating painful
348 revue neurologique 176 (2020) 325–352

diabetic neuropathy. Anesthesiol Pain Med 2015;5:e29280. randomized, sham-controlled trial. J Pain 2016;17:911–8.
http://dx.doi.org/10.5812/aapm.29280. http://dx.doi.org/10.1016/j.jpain.2016.05.003.
[95] Xu G, Xú G, Feng Y, Tang WZ, Lv ZW. Transcutaneous [107] Pei Q, Wu B, Tang Y, Yang X, Song L, Wang N, et al.
electrical nerve stimulation in combination with Repetitive transcranial magnetic stimulation at different
cobalamin injection for postherpetic neuralgia: a single- frequencies for postherpetic neuralgia: a double-blind,
center randomized controlled trial. Am J Phys Med Rehabil sham-controlled, randomized trial. Pain Physician
2014;93:287–98. http://dx.doi.org/10.1097/ 2019;22:E303–13.
PHM.0000000000000002. [108] Onesti E, Gabriele M, Cambieri C, Ceccanti M, Raccah R, Di
[96] Weintraub MI, Wolfe GI, Barohn RA, Cole SP, Parry GJ, Stefano G, et al. H-coil repetitive transcranial magnetic
Hayat G, et al. Static magnetic field therapy for stimulation for pain relief in patients with diabetic
symptomatic diabetic neuropathy: a randomized, double- neuropathy. Eur J Pain Lond Engl 2013;17:1347–56. http://
blind, placebo-controlled trial. Arch Phys Med Rehabil dx.doi.org/10.1002/j.1532-2149.2013.00320.x.
2003;84:736–46. [109] de Oliveira RAA, de Andrade DC, Mendonça M, Barros R,
[97] Wróbel MP, Szymborska-Kajanek A, Wystrychowski G, Luvisoto T, Myczkowski ML, et al. Repetitive transcranial
Biniszkiewicz T, Sieroń-Stołtny K, Sieroń A, et al. Impact of magnetic stimulation of the left premotor/dorsolateral
low frequency pulsed magnetic fields on pain intensity, prefrontal cortex does not have analgesic effect on central
quality of life and sleep disturbances in patients with poststroke pain. J Pain 2014;15:1271–81. http://dx.doi.org/
painful diabetic polyneuropathy. Diabetes Metab 10.1016/j.jpain.2014.09.009.
2008;34:349–54. http://dx.doi.org/10.1016/ [110] Galhardoni R, Aparecida da Silva V, Garcı́a-Larrea L, Dale
j.diabet.2008.02.003. C, Baptista AF, Barbosa LM, et al. Insular and anterior
[98] Weintraub MI, Herrmann DN, Smith AG, Backonja MM, cingulate cortex deep stimulation for central neuropathic
Cole SP. Pulsed electromagnetic fields to reduce diabetic pain: disassembling the percept of pain. Neurology
neuropathic pain and stimulate neuronal repair: a 2019;92:e2165–7. http://dx.doi.org/10.1212/
randomized controlled trial. Arch Phys Med Rehabil WNL.0000000000007396.
2009;90:1102–9. http://dx.doi.org/10.1016/ [111] Mori F, Codecà C, Kusayanagi H, Monteleone F, Buttari F,
j.apmr.2009.01.019. Fiore S, et al. Effects of anodal transcranial direct current
[99] Bosi E, Conti M, Vermigli C, Cazzetta G, Peretti E, Cordoni stimulation on chronic neuropathic pain in patients with
MC, et al. Effectiveness of frequency-modulated multiple sclerosis. J Pain 2010;11:436–42. http://dx.doi.org/
electromagnetic neural stimulation in the treatment of 10.1016/j.jpain.2009.08.011.
painful diabetic neuropathy. Diabetologia 2005;48:817–23. [112] Wrigley PJ, Gustin SM, McIndoe LN, Chakiath RJ,
http://dx.doi.org/10.1007/s00125-005-1734-2. Henderson LA, Siddall PJ. Longstanding neuropathic pain
[100] Bosi E, Bax G, Scionti L, Spallone V, Tesfaye S, Valensi P, after spinal cord injury is refractory to transcranial direct
et al. Frequency-modulated electromagnetic neural current stimulation: a randomized controlled trial. Pain
stimulation (FREMS) as a treatment for symptomatic 2013;154:2178–84. http://dx.doi.org/10.1016/
diabetic neuropathy: results from a double-blind, j.pain.2013.06.045.
randomised, multicentre, long-term, placebo-controlled [113] Lewis GN, Rice DA, Kluger M, McNair PJ. Transcranial
clinical trial. Diabetologia 2013;56:467–75. http:// direct current stimulation for upper limb neuropathic
dx.doi.org/10.1007/s00125-012-2795-7. pain: a double-blind randomized controlled trial. Eur J
[101] Kang BS, Shin HI, Bang MS. Effect of repetitive transcranial Pain Lond Engl 2018;22:1312–20. http://dx.doi.org/10.1002/
magnetic stimulation over the hand motor cortical area ejp.1220.
on central pain after spinal cord injury. Arch Phys Med [114] Berra E, Bergamaschi R, De Icco R, Dagna C, Perrotta A,
Rehabil 2009;90:1766–71. http://dx.doi.org/10.1016/ Rovaris M, et al. The effects of transcutaneous spinal
j.apmr.2009.04.008. direct current stimulation on neuropathic pain in multiple
[102] Ahmed MA, Mohamed SA, Sayed D. Long-term antalgic sclerosis: clinical and neurophysiological assessment.
effects of repetitive transcranial magnetic stimulation of Front Hum Neurosci 2019;13:31. http://dx.doi.org/10.3389/
motor cortex and serum beta-endorphin in patients with fnhum.2019.00031.
phantom pain. Neurol Res 2011;33:953–8. http://dx.doi.org/ [115] Tan G, Rintala DH, Thornby JI, Yang J, Wade W, Vasilev C.
10.1179/1743132811Y.0000000045. Using cranial electrotherapy stimulation to treat pain
[103] Hosomi K, Shimokawa T, Ikoma K, Nakamura Y, associated with spinal cord injury. J Rehabil Res Dev
Sugiyama K, Ugawa Y, et al. Daily repetitive transcranial 2006;43:461–74.
magnetic stimulation of primary motor cortex for [116] Tan G, Rintala DH, Jensen MP, Richards JS, Holmes SA,
neuropathic pain: a randomized, multicenter, double- Parachuri R, et al. Efficacy of cranial electrotherapy
blind, crossover, sham-controlled trial. Pain stimulation for neuropathic pain following spinal cord
2013;154:1065–72. http://dx.doi.org/10.1016/ injury: a multi-site randomized controlled trial with a
j.pain.2013.03.016. secondary 6-month open-label phase. J Spinal Cord Med
[104] Khedr EM, Kotb HI, Mostafa MG, Mohamad MF, Amr 2011;34:285–96. http://dx.doi.org/10.1179/
SA, Ahmed MA, et al. Repetitive transcranial magnetic 2045772311Y.0000000008.
stimulation in neuropathic pain secondary to [117] Chang MC. Efficacy of pulsed radiofrequency stimulation
malignancy: a randomized clinical trial. Eur J Pain in patients with peripheral neuropathic pain: a narrative
Lond Engl 2015;19:519–27. http://dx.doi.org/10.1002/ review. Pain Physician 2018;21:E225–34.
ejp.576. [118] Geurts JWM, van Wijk RMAW, Wynne HJ, Hammink E,
[105] Ma S-M, Ni J-X, Li X-Y, Yang L-Q, Guo Y-N, Tang Y-Z. High- Buskens E, Lousberg R, et al. Radiofrequency lesioning of
frequency repetitive transcranial magnetic stimulation dorsal root ganglia for chronic lumbosacral radicular pain:
reduces pain in postherpetic neuralgia. Pain Med Malden a randomised, double-blind, controlled trial. Lancet Lond
Mass 2015;16:2162–70. http://dx.doi.org/10.1111/ Engl 2003;361:21–6.
pme.12832. [119] Van Zundert J, Patijn J, Kessels A, Lamé I, van Suijlekom H,
[106] Malavera A, Silva FA, Fregni F, Carrillo S, Garcia RG. van Kleef M. Pulsed radiofrequency adjacent to the
Repetitive transcranial magnetic stimulation for phantom cervical dorsal root ganglion in chronic cervical radicular
limb pain in land mine victims: a double-blinded, pain: a double-blind sham-controlled randomized clinical
revue neurologique 176 (2020) 325–352 349

trial. Pain 2007;127:173–82. http://dx.doi.org/10.1016/ randomized comparative trial. Pain 2017;158:669–81.


j.pain.2006.09.002. http://dx.doi.org/10.1097/j.pain.0000000000000814.
[120] Simopoulos TT, Kraemer J, Nagda JV, Aner M, Bajwa ZH. [133] North RB, Kidd DH, Farrokhi F, Piantadosi SA. Spinal cord
Response to pulsed and continuous radiofrequency stimulation versus repeated lumbosacral spine surgery for
lesioning of the dorsal root ganglion and segmental nerves chronic pain: a randomized, controlled trial. Neurosurgery
in patients with chronic lumbar radicular pain. Pain 2005;56:98–106. http://dx.doi.org/10.1227/
Physician 2008;11:137–44. 01.neu.0000144839.65524.e0 [discussion 106–107].
[121] Ke M, Yinghui F, Yi J, Xeuhua H, Xiaoming L, Zhijun C, [134] Kumar K, Taylor RS, Jacques L, Eldabe S, Meglio M, Molet J,
et al. Efficacy of pulsed radiofrequency in the treatment of et al. Spinal cord stimulation versus conventional medical
thoracic postherpetic neuralgia from the angulus costae: a management for neuropathic pain: a multicentre
randomized, double-blinded, controlled trial. Pain randomised controlled trial in patients with failed back
Physician 2013;16:15–25. surgery syndrome. Pain 2007;132:179–88. http://dx.doi.org/
[122] Shanthanna H, Chan P, McChesney J, Thabane L, Paul J. 10.1016/j.pain.2007.07.028.
Pulsed radiofrequency treatment of the lumbar dorsal root [135] de Vos CC, Meier K, Zaalberg PB, Nijhuis HJA, Duyvendak
ganglion in patients with chronic lumbar radicular pain: a W, Vesper J, et al. Spinal cord stimulation in patients with
randomized, placebo-controlled pilot study. J Pain Res painful diabetic neuropathy: a multicentre randomized
2014;7:47–55. http://dx.doi.org/10.2147/JPR.S55749. clinical trial. Pain 2014;155:2426–31. http://dx.doi.org/
[123] Koh W, Choi S-S, Karm MH, Suh JH, Leem JG, Lee JD, et al. 10.1016/j.pain.2014.08.031.
Treatment of chronic lumbosacral radicular pain using [136] Slangen R, Schaper NC, Faber CG, Joosten EA, Dirksen CD,
adjuvant pulsed radiofrequency: a randomized controlled van Dongen RT, et al. Spinal cord stimulation and pain
study. Pain Med Malden Mass 2015;16:432–41. http:// relief in painful diabetic peripheral neuropathy: a
dx.doi.org/10.1111/pme.12624. prospective two-center randomized controlled trial.
[124] Makharita MY, Amr YM. Pulsed radiofrequency for Diabetes Care 2014;37:3016–24. http://dx.doi.org/10.2337/
chronic inguinal neuralgia. Pain Physician 2015;18:E147– dc14-0684.
55. [137] Kapural L, Yu C, Doust MW, Gliner BE, Vallejo R, Sitzman
[125] Chang MC, Cho YW, Ahn SH. Comparison between bipolar BT, et al. Novel 10-kHz high-frequency therapy (HF10
pulsed radiofrequency and monopolar pulsed Therapy) is superior to traditional low-frequency spinal
radiofrequency in chronic lumbosacral radicular pain: a cord stimulation for the treatment of chronic back and leg
randomized controlled trial. Medicine (Baltimore) pain: the SENZA-RCT randomized controlled trial.
2017;96:e6236. http://dx.doi.org/10.1097/ Anesthesiology 2015;123:851–60. http://dx.doi.org/10.1097/
MD.0000000000006236. ALN.0000000000000774.
[126] Halim W, van der Weegen W, Lim T, Wullems JA, Vissers [138] Bolash R, Creamer M, Rauck R, Vahedifar P, Calodney A,
KC. Percutaneous cervical nucleoplasty vs. pulsed radio Fox I, et al. Wireless high-frequency spinal cord
frequency of the dorsal root ganglion in patients with stimulation (10 kHz) compared with multiwaveform low-
contained cervical disk herniation; a prospective, frequency spinal cord stimulation in the management of
randomized controlled trial. Pain Pract 2017;17:729–37. chronic pain in failed back surgery syndrome subjects:
http://dx.doi.org/10.1111/papr.12517. preliminary results of a multicenter, prospective
[127] Luo F, Wang T, Shen Y, Meng L, Lu J, Ji N. High voltage randomized controlled study. Pain Med 2019;20(10):1971–
pulsed radiofrequency for the treatment of refractory 9. http://dx.doi.org/10.1093/pm/pnz019.
neuralgia of the infraorbital nerve: a prospective double- [139] Al-Kaisy A, Palmisani S, Pang D, Sanderson K, Wesley S,
blinded randomized controlled study. Pain Physician Tan Y, et al. Prospective, randomized, sham-control,
2017;20:271–9. double-blind, crossover trial of subthreshold spinal cord
[128] Wang F, Zhou Q, Xiao L, Yang J, Xong D, Li D, et al. A stimulation at various kilohertz frequencies in subjects
randomized comparative study of pulsed radiofrequency suffering from failed back surgery syndrome (SCS
treatment with or without selective nerve root block for Frequency Study). Neuromodulation J Int
chronic cervical radicular pain. Pain Pract 2017;17:589–95. Neuromodulation Soc 2018;21:457–65. http://dx.doi.org/
http://dx.doi.org/10.1111/papr.12493. 10.1111/ner.12771.
[129] Makharita MY, El Bendary HM, Sonbul ZM, Ahmed SES, [140] Lefaucheur J-P, Drouot X, Cunin P, Bruckert R, Lepetit H,
Latif MA. Ultrasound-guided pulsed radiofrequency in the Créange A, et al. Motor cortex stimulation for the
management of thoracic postherpetic neuralgia: a treatment of refractory peripheral neuropathic pain. Brain
randomized, double-blinded, controlled trial. Clin J Pain J Neurol 2009;132:1463–71. http://dx.doi.org/10.1093/brain/
2018;34:1017–24. http://dx.doi.org/10.1097/ awp035.
AJP.0000000000000629. [141] Robert R, Labat J-J, Bensignor M, Glemain P, Deschamps C,
[130] Hamza MA, White PF, Craig WF, Ghoname ES, Ahmed HE, Raoul S, et al. Decompression and transposition of the
Proctor TJ, et al. Percutaneous electrical nerve stimulation: pudendal nerve in pudendal neuralgia: a randomized
a novel analgesic therapy for diabetic neuropathic pain. controlled trial and long-term evaluation. Eur Urol
Diabetes Care 2000;23:365–70. 2005;47:403–8. http://dx.doi.org/10.1016/
[131] Deer T, Pope J, Benyamin R, Vallejo R, Friedman A, j.eururo.2004.09.003.
Caraway D, et al. Prospective, multicenter, randomized, [142] Gerdesmeyer L, Wagenpfeil S, Birkenmaier C, Veihelmann
double-blinded, partial crossover study to assess the A, Hauschild M, Wagner K, et al. Percutaneous epidural
safety and efficacy of the novel neuromodulation system lysis of adhesions in chronic lumbar radicular pain: a
in the treatment of patients with chronic pain of randomized, double-blind, placebo-controlled trial. Pain
peripheral nerve origin. Neuromodulation J Int Physician 2013;16:185–96.
Neuromodulation Soc 2016;19:91–100. http://dx.doi.org/ [143] Macaré van Maurik JFM, van Hal M, van Eijk RPA, Kon M,
10.1111/ner.12381. Peters EJG. Value of surgical decompression of
[132] Deer TR, Levy RM, Kramer J, Poree L, Amirdelfan K, Grigsby compressed nerves in the lower extremity in patients with
E, et al. Dorsal root ganglion stimulation yielded higher painful diabetic neuropathy: a randomized controlled
treatment success rate for complex regional pain trial. Plast Reconstr Surg 2014;134:325–32. http://
syndrome and causalgia at 3 and 12 months: a dx.doi.org/10.1097/PRS.0000000000000369.
350 revue neurologique 176 (2020) 325–352

[144] Lindblad K, Bergkvist L, Johansson A-C. Evaluation of the et al. Efficacy of oral mixed tocotrienols in diabetic
treatment of chronic chemotherapy-induced peripheral peripheral neuropathy: a randomized clinical trial. JAMA
neuropathy using long-wave diathermy and interferential Neurol 2018;75:444–52. http://dx.doi.org/10.1001/
currents: a randomized controlled trial. Support Care jamaneurol.2017.4609.
Cancer 2016;24:2523–31. http://dx.doi.org/10.1007/s00520- [157] Toth C, Brady S, Gagnon F, Wigglesworth K. A
015-3060-7. randomized, single-blind, controlled, parallel assignment
[145] Sindou M, Mertens P, Wael M. Microsurgical DREZotomy study of exercise versus education as adjuvant in the
for pain due to spinal cord and/or cauda equina injuries: treatment of peripheral neuropathic pain. Clin J Pain
long-term results in a series of 44 patients. Pain 2014;30:111–8. http://dx.doi.org/10.1097/
2001;92:159–71. http://dx.doi.org/10.1016/s0304- AJP.0b013e31828ccd0f.
3959(00)00487-5. [158] Barbin J, Seetha V, Casillas JM, Paysant J, Pérennou D. The
[146] Aichaoui F, Mertens P, Sindou M. Dorsal root entry zone effects of mirror therapy on pain and motor control of
lesioning for pain after brachial plexus avulsion: results phantom limb in amputees: a systematic review. Ann
with special emphasis on differential effects on the Phys Rehabil Med 2016;59:270–5. http://dx.doi.org/10.1016/
paroxysmal versus the continuous components. A j.rehab.2016.04.001.
prospective study in a 29-patient consecutive series. Pain [159] Thompson T, Terhune DB, Oram C, Sharangparni J, Rouf R,
2011;152:1923–30. http://dx.doi.org/10.1016/ Solmi M, et al. The effectiveness of hypnosis for pain
j.pain.2011.03.037. relief: a systematic review and meta-analysis of 85
[147] Evans S, Fishman B, Spielman L, Haley A. Randomized controlled experimental trials. Neurosci Biobehav Rev
trial of cognitive behavior therapy versus supportive 2019;99:298–310. http://dx.doi.org/10.1016/
psychotherapy for HIV-related peripheral neuropathic j.neubiorev.2019.02.013.
pain. Psychosomatics 2003;44:44–50. http://dx.doi.org/ [160] Vickers AJ, Vertosick EA, Lewith G, MacPherson H, Foster
10.1176/appi.psy.44.1.44. NE, Sherman KJ, et al. Acupuncture for chronic pain:
[148] Heutink M, Post MWM, Bongers-Janssen HMH, Dijkstra update of an individual patient data meta-analysis. J Pain
CA, Snoek GJ, Spijkerman DCM, et al. The CONECSI trial: 2018;19:455–74. http://dx.doi.org/10.1016/
results of a randomized controlled trial of a j.jpain.2017.11.005.
multidisciplinary cognitive behavioral program for coping [161] Brown S, Johnston B, Amaria K, Watkins J, Campbell F,
with chronic neuropathic pain after spinal cord injury. Pehora C, et al. A randomized controlled trial of
Pain 2012;153:120–8. http://dx.doi.org/10.1016/ amitriptyline versus gabapentin for complex regional pain
j.pain.2011.09.029. syndrome type I and neuropathic pain in children. Scand J
[149] Knoerl R, Smith EML, Barton DL, Williams DA, Holden JE, Pain 2016;13:156–63. http://dx.doi.org/10.1016/
Krauss JC, et al. Self-guided online cognitive behavioral j.sjpain.2016.05.039.
strategies for chemotherapy-induced peripheral [162] Lebrun C, Vukusic S, French Group for Recommendations
neuropathy: a multicenter, pilot, randomized, wait-list in Multiple Sclerosis (France4MS), the Société
controlled trial. J Pain 2018;19:382–94. http://dx.doi.org/ francophone de la sclérose en plaques (SFSEP).
10.1016/j.jpain.2017.11.009. Immunization and multiple sclerosis: recommendations
[150] Meize-Grochowski R, Shuster G, Boursaw B, DuVal M, from the French Multiple Sclerosis Society. Rev Neurol
Murray-Krezan C, Schrader R, et al. Mindfulness (Paris) 2019;175:341–57. http://dx.doi.org/10.1016/
meditation in older adults with postherpetic neuralgia: a j.neurol.2019.04.001.
randomized controlled pilot study. Geriatr Nurs N Y N [163] Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, et al.
2015;36:154–60. http://dx.doi.org/10.1016/ GRADE guidelines: 1. Introduction-GRADE evidence
j.gerinurse.2015.02.012. profiles and summary of findings tables. J Clin Epidemiol
[151] Nathan HJ, Poulin P, Wozny D, Taljaard M, Smyth C, Gilron 2011;64:383–94. http://dx.doi.org/10.1016/
I, et al. Randomized trial of the effect of mindfulness- j.jclinepi.2010.04.026.
based stress reduction on pain-related disability, pain [164] Attal N, Fermanian C, Fermanian J, Lanteri-Minet M,
intensity, health-related quality of life, and A1C in Alchaar H, Bouhassira D. Neuropathic pain: are there
patients with painful diabetic peripheral neuropathy. Clin distinct subtypes depending on the aetiology or
Diabetes Publ Am Diabetes Assoc 2017;35:294–304. http:// anatomical lesion? Pain 2008;138:343–53. http://dx.doi.org/
dx.doi.org/10.2337/cd17-0077. 10.1016/j.pain.2008.01.006.
[152] Gok Metin Z, Arikan Donmez A, Izgu N, Ozdemir L, Arslan [165] Baron R, Maier C, Attal N, Binder A, Bouhassira D, Cruccu
IE. Aromatherapy massage for neuropathic pain and G, et al. Peripheral neuropathic pain: a mechanism-related
quality of life in diabetic patients. J Nurs Scholarsh organizing principle based on sensory profiles. Pain
2017;49:379–88. http://dx.doi.org/10.1111/jnu.12300. 2017;158:261–72. http://dx.doi.org/10.1097/
[153] Estores I, Chen K, Jackson B, Lao L, Gorman PH. Auricular j.pain.0000000000000753.
acupuncture for spinal cord injury related neuropathic [166] Baron R, Mayoral V, Leijon G, Binder A, Steigerwald I,
pain: a pilot controlled clinical trial. J Spinal Cord Med Serpell M. 5% lidocaine-medicated plaster versus
2017;40:432–8. http://dx.doi.org/10.1080/ pregabalin in post-herpetic neuralgia and diabetic
10790268.2016.1141489. polyneuropathy: an open-label, non-inferiority two-stage
[154] Anwar S, Mowafy Z, Ali Z, Hamed H. Visual Analogue and RCT study. Curr Med Res Opin 2009;25:1663–76. http://
Neuropathic Pain Scales response to low level laser in dx.doi.org/10.1185/03007990903047880.
cases of postherpetic neuralgia of the sciatic nerve. Int J [167] Haanpää M, Cruccu G, Nurmikko TJ, McBride WT, Docu
PharmTech Res 2016;9:30–6. Axelarad A, Bosilkov A, et al. Capsaicin 8% patch versus
[155] Rajanandh MG, Kosey S, Prathiksha G. Assessment of oral pregabalin in patients with peripheral neuropathic
antioxidant supplementation on the neuropathic pain pain. Eur J Pain Lond Engl 2016;20:316–28. http://
score and quality of life in diabetic neuropathy patients – a dx.doi.org/10.1002/ejp.731.
randomized controlled study. Pharmacol Rep 2014;66:44– [168] Tesfaye S, Wilhelm S, Lledo A, Schacht A, Tölle T,
8. http://dx.doi.org/10.1016/j.pharep.2013.08.003. Bouhassira D, et al. Duloxetine and pregabalin: high-dose
[156] Vitamin E in Neuroprotection Study (VENUS) monotherapy or their combination? The ‘‘COMBO-DN
Investigators, Hor CP, Fung WY, Ang HA, Lim SC, Kam LY, study’’ – a multinational, randomized, double-blind,
revue neurologique 176 (2020) 325–352 351

parallel-group study in patients with diabetic peripheral morphine for severe neuropathic or mixed pain in
neuropathic pain. Pain 2013;154:2616–25. http:// children from age 3 months to 18 years – evaluation of the
dx.doi.org/10.1016/j.pain.2013.05.043. safety, pharmacokinetics, and efficacy of a new
[169] Gomes T, Juurlink DN, Antoniou T, Mamdani MM, gabapentin liquid formulation: study protocol for a
Paterson JM, van den Brink W. Gabapentin, opioids, and randomized controlled trial. Trials 2019;20:49. http://
the risk of opioid-related death: a population-based dx.doi.org/10.1186/s13063-018-3169-3.
nested case-control study. PLoS Med 2017;14:e1002396. [184] Kaguelidou F, Le Roux E, Mangiarini L, Lundin R, de
http://dx.doi.org/10.1371/journal.pmed.1002396. Leeuw TG, Della Pasqua O, et al. Non-inferiority double-
[170] Mathieson S, Maher CG, McLachlan AJ, Latimer J, Koes BW, blind randomised controlled trial comparing gabapentin
Hancock MJ, et al. Trial of pregabalin for acute and chronic versus tramadol for the treatment of chronic
sciatica. N Engl J Med 2017;376:1111–20. http://dx.doi.org/ neuropathic or mixed pain in children and adolescents:
10.1056/NEJMoa1614292. the GABA-1 trial – a study protocol. BMJ Open
[171] Evoy KE, Morrison MD, Saklad SR. Abuse and misuse of 2019;9:e023296. http://dx.doi.org/10.1136/bmjopen-
pregabalin and gabapentin. Drugs 2017;77:403–26. http:// 2018-023296.
dx.doi.org/10.1007/s40265-017-0700-x. [185] Pickering G, Marcoux M, Chapiro S, David L, Rat P, Michel
[172] Mick G, Gillet D, Heritier C, Garcia-Porra C, Bochet E. M, et al. An algorithm for neuropathic pain management
[Prescription and uses of pregabalin: analysis of a cohort in older people. Drugs Aging 2016;33:575–83. http://
of 419 patients at a pain study and treatment centre, and dx.doi.org/10.1007/s40266-016-0389-7.
literature review]. Douleur Analg 2019;32:87–94. [186] Capriz F, Chapiro S, David L, Floccia M, Guillaumé C, Moel
[173] Molero Y, Larsson H, D’Onofrio BM, Sharp DJ, Fazel S. V, et al. Consensus multidisciplinaire d’experts en douleur
Associations between gabapentinoids and suicidal et gériatrie : utilisation des antalgiques dans la prise en
behaviour, unintentional overdoses, injuries, road traffic charge de la douleur de la personne âgée (hors
incidents, and violent crime: population-based cohort anesthésie). Douleurs Eval - Diagn - Trait 2017;18:234–47.
study in Sweden. BMJ 2019;365:l2147. http://dx.doi.org/ [187] Sultana J, Spina E, Trifirò G. Antidepressant use in the
10.1136/bmj.l2147. elderly: the role of pharmacodynamics and
[174] Crossin R, Scott D, Arunogiri S, Smith K, Dietze PM, pharmacokinetics in drug safety. Expert Opin Drug Metab
Lubman DI. Pregabalin misuse-related ambulance Toxicol 2015;11:883–92. http://dx.doi.org/10.1517/
attendances in Victoria, 2012–2017: characteristics of 17425255.2015.1021684.
patients and attendances. Med J Aust 2019;210:75–9. [188] Finnerup NB, Haroutounian S, Baron R, Dworkin RH,
http://dx.doi.org/10.5694/mja2.12036. Gilron I, Haanpaa M, et al. Neuropathic pain clinical trials:
[175] Driot D, Jouanjus E, Oustric S, Dupouy J, Lapeyre-Mestre M. factors associated with decreases in estimated drug
Patterns of gabapentin and pregabalin use and misuse: efficacy. Pain 2018;159:2339–46. http://dx.doi.org/10.1097/
results of a population-based cohort study in France. Br J j.pain.0000000000001340.
Clin Pharmacol 2019;85:1260–9. http://dx.doi.org/10.1111/ [189] Cohen J. Statistical power analysis for the behavioral
bcp.13892. sciences. 2nd ed. L. Erlbaum Associates, New-Jersey, USA;
[176] Nahar LK, Murphy KG, Paterson S. Misuse and mortality 1988.
related to gabapentin and pregabalin are being under- [190] Wilkinson ST, Ballard ED, Bloch MH, Mathew SJ, Murrough
estimated: a two-year post-mortem population study. J JW, Feder A, et al. The effect of a single dose of
Anal Toxicol 2019;43(7):564–70. http://dx.doi.org/10.1093/ intravenous ketamine on suicidal ideation: a systematic
jat/bkz036. review and individual participant data meta-analysis. Am
[177] Moisset X, Trouvin A-P, Tran V-T, Authier N, Vergne-Salle J Psychiatry 2018;175:150–8. http://dx.doi.org/10.1176/
P, Piano V, et al. [Use of strong opioids in chronic non- appi.ajp.2017.17040472.
cancer pain in adults. Evidence-based recommendations [191] Kastrup J, Petersen P, Dejgård A, Angelo HR, Hilsted J.
from the French Society for the Study and Treatment of Intravenous lidocaine infusion – a new treatment of
Pain]. Presse Med 2016;45:447–62. http://dx.doi.org/ chronic painful diabetic neuropathy? Pain 1987;28:69–75.
10.1016/j.lpm.2016.02.014. [192] Attal N, Rouaud J, Brasseur L, Chauvin M, Bouhassira D.
[178] ANSM. État des lieux de la consommation des antalgiques Systemic lidocaine in pain due to peripheral nerve injury
opioı̈des et leurs usages problématiques; 2019 [accessed and predictors of response. Neurology 2004;62:218–25.
July 19, 2019]https://ansm.sante.fr/S-informer/Points-d- http://dx.doi.org/10.1212/01.wnl.0000103237.62009.77.
information-Points-d-information/Antalgiques-opioides- [193] Tremont-Lukats IW, Hutson PR, Backonja M-M. A
l-ANSM-publie-un-etat-des-lieux-de-la-consommation- randomized, double-masked, placebo-controlled pilot trial
en-France-Point-d-Information. of extended IV lidocaine infusion for relief of ongoing
[179] Stoicea N, Costa A, Periel L, Uribe A, Weaver T, Bergese SD. neuropathic pain. Clin J Pain 2006;22:266–71. http://
Current perspectives on the opioid crisis in the US dx.doi.org/10.1097/01.ajp.0000169673.57062.40.
healthcare system: a comprehensive literature review. [194] Liu H, Lu F, Zhou D, Yin Y, Li J, Yang B, et al. The analgesic
Medicine (Baltimore) 2019;98:e15425. http://dx.doi.org/ and emotional response to intravenous lidocaine infusion
10.1097/MD.0000000000015425. in the treatment of postherpetic neuralgia: a randomized,
[180] Chenaf C, Kaboré J-L, Delorme J, Pereira B, Mulliez A, double-blinded, placebo-controlled study. Clin J Pain
Zenut M, et al. Prescription opioid analgesic use in France: 2018;34:1025–31. http://dx.doi.org/10.1097/
trends and impact on morbidity-mortality. Eur J Pain Lond AJP.0000000000000623.
Engl 2019;23:124–34. http://dx.doi.org/10.1002/ejp.1291. [195] Mhalla A, Baudic S, Ciampi de Andrade D, Gautron M,
[181] ANSM. Cannabis à visée thérapeutique en France; 2019. Perrot S, Teixeira MJ, et al. Long-term maintenance of the
[182] Boulkedid R, Abdou AY, Desselas E, Monégat M, de Leeuw analgesic effects of transcranial magnetic stimulation in
TG, Avez-Couturier J, et al. The research gap in chronic fibromyalgia. Pain 2011;152:1478–85. http://dx.doi.org/
paediatric pain: a systematic review of randomised 10.1016/j.pain.2011.01.034.
controlled trials. Eur J Pain Lond Engl 2018;22:261–71. [196] Hodaj H, Alibeu J-P, Payen J-F, Lefaucheur J-P. Treatment
http://dx.doi.org/10.1002/ejp.1137. of chronic facial pain including cluster headache by
[183] de Leeuw TG, Mangiarini L, Lundin R, Kaguelidou F, van repetitive transcranial magnetic stimulation of the motor
der Zanden T, Pasqua OD, et al. Gabapentin as add-on to cortex with maintenance sessions: a naturalistic study.
352 revue neurologique 176 (2020) 325–352

Brain Stimulat 2015;8:801–7. http://dx.doi.org/10.1016/ [206] Hosomi K, Saitoh Y, Kishima H, Oshino S, Hirata M, Tani
j.brs.2015.01.416. N, et al. Electrical stimulation of primary motor cortex
[197] Kobayashi M, Fujimaki T, Mihara B, Ohira T. Repetitive within the central sulcus for intractable neuropathic pain.
transcranial magnetic stimulation once a week induces Clin Neurophysiol 2008;119:993–1001. http://dx.doi.org/
sustainable long-term relief of central poststroke pain. 10.1016/j.clinph.2007.12.022.
Neuromodulation J Int Neuromodulation Soc 2015;18:249– [207] Lefaucheur J-P, Ménard-Lefaucheur I, Goujon C, Keravel Y,
54. http://dx.doi.org/10.1111/ner.12301. Nguyen J-P. Predictive value of rTMS in the identification
[198] Quesada C, Pommier B, Fauchon C, Bradley C, Créac’h C, of responders to epidural motor cortex stimulation
Vassal F, et al. Robot-guided neuronavigated repetitive therapy for pain. J Pain 2011;12:1102–11. http://dx.doi.org/
transcranial magnetic stimulation (rTMS) in central 10.1016/j.jpain.2011.05.004.
neuropathic pain. Arch Phys Med Rehabil 2018. http:// [208] André-Obadia N, Mertens P, Lelekov-Boissard T, Afif A,
dx.doi.org/10.1016/j.apmr.2018.04.013. Magnin M, Garcia-Larrea L. Is life better after motor cortex
[199] Perruchoud C, Eldabe S, Batterham AM, Madzinga G, stimulation for pain control? Results at long-term and
Brookes M, Durrer A, et al. Analgesic efficacy of high- their prediction by preoperative rTMS. Pain Physician
frequency spinal cord stimulation: a randomized double- 2014;17:53–62.
blind placebo-controlled study. Neuromodulation Technol [209] Pommier B, Quesada C, Fauchon C, Nuti C, Vassal F,
Neural Interface 2013;16:363–9. http://dx.doi.org/10.1111/ Peyron R. Added value of multiple versus single sessions
ner.12027. of repetitive transcranial magnetic stimulation in
[200] Duarte RV, Nevitt S, McNicol E, Taylor RS, Buchser E, North predicting motor cortex stimulation efficacy for refractory
RB, et al. Systematic review and meta-analysis of placebo/ neuropathic pain. J Neurosurg 2019;130:1750–61. http://
sham-controlled randomised trials of spinal cord dx.doi.org/10.3171/2017.12.JNS171333.
stimulation for neuropathic pain. Pain 2019;160(1):24–35. [210] Khatoun A, Asamoah B, Mc Laughlin M. Investigating the
http://dx.doi.org/10.1097/j.pain.0000000000001689. feasibility of epicranial cortical stimulation using
[201] Lima MC, Fregni F. Motor cortex stimulation for chronic concentric-ring electrodes: a novel minimally invasive
pain: systematic review and meta-analysis of the neuromodulation method. Front Neurosci 2019;13:773.
literature. Neurology 2008;70:2329–37. http://dx.doi.org/ http://dx.doi.org/10.3389/fnins.2019.00773.
10.1212/01.wnl.0000314649.38527.93. [211] Hilton L, Hempel S, Ewing BA, Apaydin E, Xenakis L,
[202] Fontaine D, Hamani C, Lozano A. Efficacy and safety of Newberry S, et al. Mindfulness meditation for chronic
motor cortex stimulation for chronic neuropathic pain: pain: systematic review and meta-analysis. Ann Behav
critical review of the literature. J Neurosurg 2009;110:251– Med Publ Soc Behav Med 2017;51:199–213. http://
6. http://dx.doi.org/10.3171/2008.6.17602. dx.doi.org/10.1007/s12160-016-9844-2.
[203] Nguyen JP, Lefaucheur JP, Raoul S, Roualdes V, Péréon Y, [212] Hughes LS, Clark J, Colclough JA, Dale E, McMillan D.
Keravel Y. Chapter 40 – Motor cortex stimulation for the Acceptance and commitment therapy (ACT) for chronic
treatment of neuropathic pain. In: Krames ES, Hunter pain: a systematic review and meta-analyses. Clin J Pain
Peckham P, Rezai AR, editors. Neuromodulation. San 2017;33:552–68. http://dx.doi.org/10.1097/
Diego: Academic Press; 2009. p. 515–26. AJP.0000000000000425.
[204] Nguyen J-P, Velasco F, Brugières P, Velasco M, Keravel Y, [213] de Vries F, Bruin M, Lobatto DJ, Dekkers OM, Schoones JW,
Boleaga B, et al. Treatment of chronic neuropathic pain by van Furth WR, et al. Opioids and their endocrine effects: a
motor cortex stimulation: results of a bicentric controlled systematic review and meta-analysis. J Clin Endocrinol
crossover trial. Brain Stimulat 2008;1:89–96. http:// Metab 2019;105(3). http://dx.doi.org/10.1210/jcemdgz022.
dx.doi.org/10.1016/j.brs.2008.03.007. [214] Farrar JT, Young JP, LaMoreaux L, Werth JL, Poole RM.
[205] Rasche D, Ruppolt M, Stippich C, Unterberg A, Tronnier Clinical importance of changes in chronic pain intensity
VM. Motor cortex stimulation for long-term relief of measured on an 11-point numerical pain rating scale. Pain
chronic neuropathic pain: a 10-year experience. Pain 2001;94:149–58. http://dx.doi.org/10.1016/s0304-
2006;121:43–52. http://dx.doi.org/10.1016/ 3959(01)00349-9.
j.pain.2005.12.006.

Vous aimerez peut-être aussi