You are on page 1of 10

DIABETES/METABOLISM RESEARCH AND REVIEWS REVIEW ARTICLE

Diabetes Metab Res Rev 2011; 27: 629–638.


Published online in Wiley Online Library (wileyonlinelibrary.com) DOI: 10.1002/dmrr.1225

Painful diabetic peripheral neuropathy: consensus


recommendations on diagnosis, assessment
and management

S. Tesfaye1 Summary
L. Vileikyte2
G. Rayman3 Painful diabetic peripheral neuropathy (DPN) is common, is associated with
S. H. Sindrup4 significant reduction in quality of life and poses major treatment challenges
B. A. Perkins5 to the practising physician. Although poor glucose control and cardiovascular
M. Baconja6 risk factors have been proven to contribute to the aetiology of DPN, risk factors
A. I. Vinik7 specific for painful DPN remain unknown. A number of instruments have been
A. J. M. Boulton2∗ on behalf of
tested to assess the character, intensity and impact of painful DPN on quality
The Toronto Expert Panel on
of life, activities of daily living and mood. Management of the patient with
Diabetic Neuropathy†
DPN must be tailored to individual requirements, taking into consideration
1
Diabetes Research Unit, the co-morbidities and other factors. Pharmacological agents with proven effi-
Royal Hallamshire Hospital and cacy for painful DPN include tricyclic anti-depressants, the selective serotonin
University of Sheffield, and noradrenaline re-uptake inhibitors, anti-convulsants, opiates, membrane
Sheffield, UK stabilizers, the anti-oxidant alpha-lipoic acid and topical agents including
2 Department of Medicine, University capsaicin. Current first-line therapies for painful DPN include tricyclic anti-
of Manchester, Manchester, UK depressants, the serotonin and noradrenaline re-uptake inhibitor duloxetine
3 IpswichDiabetes Centre, and the anti-convulsants pregabalin and gabapentin. When prescribing any
Ipswich Hospital NHS Trust, of these agents, other co-morbidities and costs must be taken into account.
Ipswich, UK Second-line approaches include the use of opiates such as synthetic opioid
4 Department of Neurology, Odense tramadol, morphine and oxycodone-controlled release. There is a limited
University Hospital, Odense, literature with regard to combination treatment. In extreme cases of painful
Denmark
5 Division
DPN unresponsive to pharmacotherapy, occasional use of electrical spinal
of Endocrinology, cord stimulation might be indicated. There are a number of unmet needs in
University of Toronto,
Toronto, ON, Canada the therapeutic management of painful DPN. These include the need for ran-
6 Department domized controlled trials with active comparators and data on the long-term
of Neurology,
University of Madison-Wisconsin, efficacy of agents used, as most trials have lasted for less than 6 months.
Madison, WI, USA Finally, there is a need for appropriately designed studies to investigate
7 Strelitz
Diabetes Research Institute, non-pharmacological approaches. Copyright  2011 John Wiley & Sons, Ltd.
Eastern Virginia Medical School,
Norfolk, VA, USA Keywords diabetic neuropathy; painful diabetic peripheral neuropathy;

Correspondence to: treatment
A. J. M. Boulton,
Manchester Diabetes Centre, Abbreviations DPN – diabetic peripheral neuropathy; HADS – Hospital
193 Hathersage Road, Anxiety and Depression Scale; QoL – quality of life; SNRIs – serotonin and
Manchester M13 OJE, UK noradrenaline re-uptake inhibitors; TCA – tricyclic anti-depressant
E-mail: ABoulton@med.miami.edu

† See Appendix for Members of The

Toronto Consensus Panel on Introduction


Diabetic Neuropathy.
This review by a group of Toronto Expert Panel on Diabetic Neuropathy
Received : 6 April 2011
examined the recent literature on painful diabetic peripheral neuropathy
Accepted: 6 June 2011 (DPN) and made consensus recommendations based on these. Although
a summarized version of this review has recently been published [1], this

Copyright  2011 John Wiley & Sons, Ltd.


630 S. Tesfaye et al.

review examines the diagnosis and pharmacological functioning over time contributed to further increments
management of painful DPN in more detail. in depressive symptoms [11]. In one study conducted in
DPN is present in up to 50% of all diabetic patients with a tertiary referral multi-disciplinary clinic for painful DPN
long duration of disease and is a major cause of morbidity comprising of patients with moderate or severe symptoms,
that is also associated with increased mortality [2–4]. over two thirds were found to have anxiety and/or depres-
Of all the neuropathies in diabetes, chronic DPN is the sion [13] and >95% had sleep disturbances [12]. With
commonest [2,5]. Up to 50% of all patients with DPN may respect to the natural history of painful DPN, there are
experience painful symptoms, although many of these will very few appropriate studies, but it is generally believed
not have pain of sufficient severity to warrant treatment. that painful symptoms may wax and wane over the years
Of all the distressing symptoms of DPN, pain is the most and eventually become less prominent as the sensory loss
prominent and most frequent reason for seeking medical worsens; however, others have suggested no appreciable
attention. A definition of neuropathic pain in diabetes occurrence of significant remissions [2].
adapted from the original International Association for
the Study of Pain’s definition [6] is ‘pain arising as a
direct consequence of abnormalities in the somatosensory Acute painful DPN
system in people with diabetes’. Patients experiencing
neuropathic pain in diabetes often find their symptoms The above discussion has referred mainly to patients
difficult to describe as they are unfamiliar, but common with the commonest variety of painful DPN, which is
descriptors include burning pain, ‘electrical shock’ type chronic distal symmetrical symptomatic polyneuropathy.
shooting pain down the legs, lancinating (often likened as The much less common though well-recognized acute
‘stabbing’ or ‘knife-like’ pains), uncomfortable tingling painful DPN is a rare yet distinct variety of symmetrical
(paresthesia or novocaine-like) and many experience polyneuropathies. It is characterized by severe sensory
contact pain brought on by touching daytime socks or symptoms similar to those described above, but with
stockings or bedclothes at night (allodynia). Discomfort few neurological signs on examination. The overriding
on walking may also be described as ‘walking barefoot symptom reported by all patients is severe pain
on marbles’ or ‘walking barefoot on hot sand’. Subjective and there may be associated weight loss, depression
sensations of altered temperature perceptions, such as and, in men, erectile dysfunction [2,14]. This acute
the feet are very warm or very cold, are also common neuropathy typically follows some rapid change in
and non-specific aching feelings in the feet and cramp- glycaemic control, which might be either following
like sensations in the legs are other frequently described sudden improvement of control such as ‘insulin neuritis’
symptoms [2,5]. Occasionally, these symptoms may [2,14] or following an episode of very poor glycaemic
progress from the feet up the limbs and involve the whole control typically in young type 1 patients such as
of the lower limbs and later there may be involvement of following diabetic ketoacidosis, with the symptoms
the upper limb, particularly the hand. It is important for coming on after successful treatment of the acute
the reader to appreciate that a host of other conditions exacerbation [2,5,14,15]. Although unfortunate, the
can masquerade as neuropathy, including entrapments, term ‘insulin neuritis’ is here to stay despite the fact
fasciitis and claudication. that it might be precipitated by sudden improvement
Neuropathic pain in diabetes is characteristically more of glycaemic control, consequent on other treatments
severe at night often resulting in sleep disturbance [7], such as oral hypoglycaemic agents. The prognosis of
with severely affected patients complaining of being con- acute painful DPN is good with complete resolution
stantly tired because of severe sleep deprivation [8]. of all symptoms usually occurring within a year of
Together with painful symptoms during the day, this onset [2,5,15].
often leads to a reduction in individuals being able to
perform daily activities. Relief of pain improves sleep and
the degree of sleep loss predicts the response to anal- Epidemiology
gesics. Pathway analysis has shown that there are both
direct and indirect actions of agents that relieve pain on Although the epidemiology and risk factors of DPN
sleep disturbances [9]. The burden of painful DPN was have been extensively studied [2,5], there are very few
reported to be considerable in one study which resulted studies that look specifically at the prevalence of pain:
in a persistent discomfort despite polypharmacy and high the prevalence varies from 10 to 26% in a number
resource use and led to limitations in daily activities and of studies [5,16]. In one population-based study in
poor satisfaction with treatments that were often deemed urban Liverpool, UK, the prevalence of painful DPN as
to be inappropriate. Chronic persistently painful DPN can assessed by a structured questionnaire and examination
be extremely distressing and might be associated with pro- was estimated at 16% [16]. Sadly, in these patients it was
found depression [10,11] together with anxiety [12] and found that 12.5% had never reported their symptoms
sleep loss. While painful DPN contributed to depression, to their doctor and notably 39% had never received
unsteadiness and its psychosocial consequences domi- treatment for their pain [16]. Similarly, it is difficult
nated this relationship over time [11]. Furthermore, a because of the lack of studies to confirm specific risk
decline in neuropathy-related physical and psychosocial factors for painful diabetic neuropathy other than the

Copyright  2011 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2011; 27: 629–638.
DOI: 10.1002/dmrr
Painful Diabetic Peripheral Neuropathy 631

well-described relationship between sudden change of As diabetic neuropathy is a diagnosis of exclusion, a


glycaemic control and acute sensory neuropathy as noted careful clinical history and a peripheral neurological
above. and vascular examination of the lower extremities are
essential to exclude other causes of neuropathic pain and
leg/foot pain such as peripheral vascular disease, arthritis,
Mechanisms of neuropathic pain in malignancy, alcohol abuse and spinal canal stenosis
diabetes [2,5,24]. As painful DPN is invariably symmetrical,
patients with asymetrical symptoms and/or signs should
The exact pathophysiological mechanisms of neuropathic be carefully assessed for other aetiologies of their
pain in diabetes remain enigmatic although several symptomatology.
mechanisms including neuro-structural correlates for A number of simple numeric rating scales can be used
painful neuropathy have been postulated (Table 1) [17]. to assess the frequency and severity of painful symptoms
Other potential mechanisms include the association [25]. It is important to emphasize the difficulties in
of increased blood glucose instability in the genesis the description and assessment of painful symptoms
of neuropathic pain [18], an increase in peripheral that patients experience: pain is a very individual
nerve epineurial blood flow [19], altered foot skin sensation and patients with similar pathological lesions
microcirculation [20], reduced intra-epidermal nerve may describe their symptoms in markedly different ways.
fibre density in the context of early neuropathy [21], It must also be remembered that as pain is a personal
increased thalamic vascularity [22] and autonomic psychological experience, the external observer can play
dysfunction [23]. no part in its assessment or interpretation. Hence, the
use of simple scales is recommended, such as simple
visual analogue style, which is of course the oldest
Assessment and diagnosis of painful and best validated measure or the numerical rating
DPN scale, such as an 11-point Likert scale (0 = no pain
to 10 = worst possible pain). Other validated scales and
As recommended by the American Diabetes Association questionnaires including the neuropathic pain symptom
Consensus Statement [24], the diagnosis of painful DPN inventory [26], the modified brief pain inventory [27],
in practice is a clinical one, relying on the patient’s the neuropathic pain questionnaire [28], the LANNS pain
description of pain: symptoms distal, symmetrical and scale [29] and the McGill Pain Questionnaire are often
associated with noctural exacerbation and commonly used used in its shortened format [30]. Quality of life (QoL)
descriptors as noted above [24]. The clinical diagnosis might be assessed using generic instruments; however,
may be supported by nerve conduction studies and an inherent limitation of these is that their content did
quantitative sensory testing. Nerve conduction studies not emerge from patients affected by neuropathic pain,
are particularly important to exclude other cases of pain, unlike neuropathy-specific instruments that are based on
e.g. entrapment syndromes. The diagnostic criteria for the patient’s experience of neuropathic pain. Thus, it
DPN (possible, probable and confirmed) are discussed in may be preferable to use validated, neuropathy-specific
Ref. [1]. On examination, there is usually blunting of QoL such as NeuroQol [31], Norfolk Quality of Life
sensation on clinical examination of the feet although Scale [32] and Neuropathic Pain Impact on Quality-
occasionally as noted above in acute painful DPN, of-Life questionnaire (NePIQoL) [33]. The impact of
symptoms may be present in the absence of signs [2,5,14]. painful symptomatology on mood can be evaluated using
scales such as the Hospital Anxiety and Depression
Table 1. Mechanisms of neuropathic pain Scale (HADS) [34]. Similarly, there are a number
of scales that might assess pain behaviour and sleep
Peripheral mechanisms Central mechanisms interference and the prediction of response to therapy
[9,15].
Changes in sodium channel Central sensitization
distribution and expression For clinical trials of putative new therapies for
Changes in calcium channel Aβ fibre sprouting into lamina painful DPN, rigorous patient selection with the use
distribution and expression II of the dorsal horn of neuropathic pain scales and outcome measures
Altered neuro-peptide Reduced inhibition via are indicated. Inclusion criteria for such trials would
expression descending pathways
normally include neuropathic pain associated with
Sympathetic sprouting
Peripheral sensitization diabetes for greater than 6 months but less than
Altered peripheral blood flow 5 years of duration, and mean weekly pain score of
Axonal atrophy, degeneration between 4 and 10 on an 11-point graphic rating
or regeneration scale, with exclusion of pain not associated with DPN,
Damage to small fibres
Glycaemic flux
those with mono or proximal neuropathies, other non-
neuropathic chronic pain and those with central causes of
Adapted from Ref. [17]. pain.

Copyright  2011 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2011; 27: 629–638.
DOI: 10.1002/dmrr
632 S. Tesfaye et al.

Management of painful DPN Table 2. Lifestyle, metabolic control and pharmacologi-


cal treatment approaches for painful diabetic peripheral
neuropathy showing some of the commonly prescribed
The assessment and treatment of painful DPN continues treatments
to pose a considerable challenge to clinicians and an
empathic and multi-disciplinary approach is crucial as the • Physiological glucose control (HbA1c 6–7%)
impact of painful DPN is varied and multi-dimensional • Lifestyle modification (diet, exercise)
• Management of cardiovascular risk factors
[5]. Ideally, a multi-disciplinary team might include
• Tricyclic anti-depressants
input from diabetologists/endocrinologists, neurologists, Amitriptyline 25–75 mg/day
the pain clinic team, specialist nurses, podiatrists, psy- Imipramine 25–75 mg/day
chologists, physiotherapists and others. However, in most • Serotonin noradrenalin re-uptake inhibitors
clinical settings this is not possible and the management Duloxetine 60–120 mg/day (indicated for painful diabetic
falls mainly to the diabetes physician, the primary care peripheral neuropathy by US Food and Drug
Administration and European Medicines Agency)
physician or neurologist. Although strong evidence impli-
Venlafaxine 150–225 mg/day
cates poor glycaemic control as a pathogenetic mechanism • Anti-convulsants
in the aetiology of DPN, there is no proof from randomized Gabapentin 900–3600 mg/day
controlled trials that this is the case for pain symptomatol- Pregabalin 300–600 mg/day (indicated for painful diabetic
ogy. However, a number of observational studies suggest peripheral neuropathy by US Food and Drug
that poor or erratic glycaemic control contributes to the Administration and European Medicines Agency)
Carbamazepine 200–800 mg/day
genesis of neuropathic pain [2] and there is also some evi-
Topiramate 25–100 mg/day
dence that increased blood glucose flux might contribute • Opiates
to neuropathic pain [18]. A randomized controlled trial in Tramadol 200–400 mg/day
this area is unlikely and there is therefore general consen- Oxycodone 20–80 mg/day
sus that good blood glucose control should be the first step Morphine sulfate sustained-release 20–80 mg/day
in the management of any form of diabetic neuropathy • Capsaicin cream
(0.075%) Applied sparingly three to four times per day
(with the caveats above on acute painful neuropathy). As
other risk factors of large vessel disease are also common
in DPN (e.g. hypertension, hyperlipidaemia), it is impor-
and more recently the antagonism of n-methyl-D-aspartate
tant to address these abnormalities in addition (with the
receptors, which mediate hyperalgesia and allodynia [36].
caveat that the use of lipid-lowering drugs can case a
Amitriptyline and imipramine have balanced inhibition of
painful neuropathic syndrome rarely).
noradrenaline and serotonin, which may be an advantage
with respect to efficacy over noradrenergic compounds
such as nortriptyline and desipramine, that on the other
Pharmacological management of hand are better tolerated. In addition, drugs such as nor-
painful DPN triptyline have less anti-cholinergic properties and cause
less of the symptoms due to cholinergic blockade. If
A large number of pharmacological treatments with carefully titrated the number needed to treat (NNT) is
known efficacy in diabetic neuropathy are listed in 1.5–3.5 [15]. However, some of the trials were small
Table 2, although only two (duloxetine and pregabalin) and crossover that may have influenced the NNTs to be
are approved for the treatment of neuropathic pain in lower. As the TCAs have predictable and frequent side
diabetes by both the Food and Drugs Administration of effects including drowsiness and anti-cholinergic effects,
the United States and the European Medicines Agency. it is recommended to start at a small dose especially
It should be noted that, with the exception of tight gly- in older patients of 10 mg/day, increasing as needed to
caemic control, none of the treatments listed in Table 2 75 mg/day. As data from a large retrospective study of
will have any effect on the natural history of DPN which patients on TCA therapy showed an increased risk of sud-
is a progressive loss of nerve fibres: all are therefore den cardiac death associated with doses of >100 mg/day
symptomatic treatments only. [37], then caution should be taken in any patient with
a history of cardiovascular disease in addition to older
patients. Some authorities recommend that an electrocar-
Tricyclic anti-depressants diogram should be carried out and if there is prolongation
of the PR or QTc interval these drugs should not be used.
Several randomized controlled trials and meta-analyses
have confirmed the efficacy of tricyclic anti-depressants
(TCAs) in painful DPN [35]. These agents have been Serotonin and noradrenalin re-uptake
shown to promote successful analgesia to thermal, inhibitors
mechanic and electrical stimuli in diabetic patients: puta-
tive mechanisms underlying these effects include the Serotonin and noradrenalin re-uptake inhibitors (SNRIs)
inhibition of noradrenaline and/or serotonin re-uptake such as duloxetine relieve pain by increasing the synaptic
synapses of central descending pain-controlled systems availability of 5-hydroxytryptamine and noradrenaline

Copyright  2011 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2011; 27: 629–638.
DOI: 10.1002/dmrr
Painful Diabetic Peripheral Neuropathy 633

in the descending pathways that are inhibitory to pain of topiramate (up to 600 mg/day) in subjects with
impulses. A further advantage of duloxetine is that it has moderately to severely painful DPN suggested that pain
anti-depressant effects in addition to the analgesic effects relief was effective and the drug caused weight loss and
in diabetic neuropathy. The efficacy of this agent has been improvement in lipid and blood pressure parameters but
confirmed in painful DPN in several similar clinical trials 39.5% of subjects discontinued, most often due to adverse
at doses of 60 or 120 mg/day [35,38]. The pooled data events [41].
from three trials confirmed that efficacy was maintained
throughout the treatment period of 12 weeks and that
approximately 50% of patients had achieved at least 50% Local anaesthetic or anti-arrhythmic
pain reduction [38].
agents
The NNT to achieve at least 50% pain reduction
(generally accepted to be clinically meaningful) was The benefit of intravenous lidocaine (5 mg/kg over
4.9 for 120 mg/day and 5.2 for 60 mg/day [38]. An 30 min) in painful DPN was confirmed in a randomized
advantage of this agent is that it is not associated with double-blind placebo controlled trial [42]. However, oral
weight gain and the most frequent adverse effects include dosing is unavailable and electrocardiogram monitoring
nausea, somnolence, dizziness, constipation, dry mouth is necessary during administration: its use is therefore
and reduced appetite, although these tend to be mild to limited to refractory cases of painful DPN.
moderate and are transient. Mexiletine is a class 1B anti-arrhythmic agent that
Another SNRI, venlafaxine (dosages 150–225 mg/day), is an orally available structural analogue of lidocaine.
is also effective in relieving painful symptomatology [35], Its efficacy in reducing neuropathic pain has been
although cardiovascular adverse events limit its use in confirmed in clinical trials, although a review of seven
diabetes. control trials suggested that mexiletine only provided a
modest analgesic effect [43]: as regular electrocardiogram
monitoring is necessary, the long-term use of mexiletine
Anti-convulsants
cannot be recommended.
Gabapentin and pregabalin, which bind to the α-2-δ A multicentre randomized, open-label, parallel-group
subunit of the calcium channel, reducing calcium influx study of lidocaine patch versus pregabalin with a drug
and thus resulting in reduced synaptic neurotransmitter washout phase of up to 2 weeks and a comparative phase
release into the hyperexcited neurone are the two anti- of 4-week treatment period showed that lidocaine was as
convulsants most frequently used to treat neuropathic effective as pregabalin in reducing pain and was free of
pain [35]. side effects [35].
The evidence for the efficacy of the first generation
agent (e.g. carbamazepine, phenytoin) is limited and
mainly comes from small single-centre studies. Further- Opioids
more, these agents are associated with a relatively high
frequency of adverse events, particularly central effects Many physicians are reluctant to prescribe opioids for
such as somnolence and dizziness [2,35]. Gabapentin is neuropathic pain probably because of fear of addiction.
well established as a treatment of painful DPN, although However, there is randomized controlled trial evidence
doses typically prescribed in clinical practice are much for some opiods and therefore there is a good rationale
lower than the doses used in the main clinical trial of up for their use in appropriate patient after trail in first-line
to 3.6 g/day [39]. therapy. Of the orally administered opioids, tramadol
The second commonly used agent in this group, is the best studied. It is a centrally acting synthetic
pregabalin, has been shown to be highly effective in opioid with an unusual mode of action, working on both
the treatment of painful DPN in several randomized opioid and mono-aminergic pathways. It has a lower
controlled trials [35,40]. Based upon these and other abuse potential than conventionally stronger opioids and
evidence supporting its efficacy and tolerability, doses of development of tolerance is uncommon. In a randomized
150–600 mg/day (in divided doses) for the treatment of controlled trial, tramadol up to 200 mg/day was effective
diabetic neuropathic pain are recommended. The pooled in the management of painful DPN with a follow-up
analysis of seven randomized controlled trials in painful showing that symptomatic relief could be maintained for
DPN has confirmed the efficacy and safety of pregabalin at least 6 months [44]. Finally, two randomized trials have
[40]. Data from this pooled analysis showed an NNT confirmed the efficacy of controlled release of oxycodone
of 4.04 for 600 mg/day and 5.99 for 300 mg/day. The for neuropathic pain in diabetes [35,45]. All studies of
most frequent side effects for pregabalin are dizziness, opioids have only assessed relatively short-term use, so
somnolence, peripheral oedema, headache and weight the risks of tolerance and dependence in longer term
gain. usage are yet to be quantified. In recognition of these
Trials on anti-convulsants lamotrigine and lacosamide potential problems, physicians should be alert to signs of
indicate some efficacy of these compounds, but the abuse and should only use the opioids if other therapies
results are equivocal [35]. An open-label extension study have failed to providing sufficient pain relief.

Copyright  2011 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2011; 27: 629–638.
DOI: 10.1002/dmrr
634 S. Tesfaye et al.

Comparative or combination trials or pregabalin as first-line approaches. They also recom-


mended that the SNRI, duloxetine, could be the first-
A major problem in the area of treatment of neuropathic or second-line treatment. Previously, guidelines from the
pain in diabetes is the relative lack of comparative or Canadian Pain Society [52] had made similar proposals
combination studies: most cited studies in this review with TCAs and anti-convulsants being the first-line treat-
have been of active agents against placebo, whereas there ments and the SNRI the second line. Finally, the European
is a need for more studies that compare a new drug Federation of Neurological Society’s Guidelines proposed
with an active comparator or indeed two existing drugs that first-line treatments might comprise of TCAs, SNRIs,
with known efficacy. A recent example of such a trial is gabapentin or pregabalin [53]. Most recently, the UK
that of Bansal et al. [46] who compared amitriptyline National Institute for Health and Clinical Excellence pub-
with pregabalin in painful diabetic neuropathy in a lished guidelines on the management of neuropathic pain
randomized double-blind trial. This study confirmed that in non-specialist settings, in which the management of
although there was little difference in efficacy, pregabalin painful DPN featured prominently [54]. While National
was the preferred drug because of a superior adverse Institute for Health and Clinical Excellence ranked the
event profile. Similarly, Gilron et al. studied nortriptyline level of evidence of pain outcomes with duloxetine, pre-
and gabapentin either in combination or alone in a gabalin and gabapentin as similar, they propose that
randomized trial and confirmed that when given together, oral duloxetine should be the first-line treatment with
they were more efficacious than either drug given alone amitriptyline as an alternative and pregabalin as a second-
[47]. In a crossover study, low-dose combination therapy line treatment [54]. The proposal that oral duloxetine
with gabapentin and morphine was significantly more should be the first-line therapy does not appear to be
effective than either monotherapy at higher doses [48]. based on efficacy but rather cost-effectiveness.

Topical treatments Recommendations for treatment


algorithm
Topical treatments offer several theoretical advantages
including minimal side effects, lack of drug interactions On the basis of the clinical trial evidence of the various
and no need for dose titration. However, few have been pharmacological agents for painful DPN, the following
evaluated in well-designed randomized control trials. treatment algorithm is recommended (Figure 1). The
Topical capsaicin works by releasing substance ‘P’ from panel compared the relative efficacy and safety of
nerve terminals which become depleted and there might treatments for painful DPN and recommended that a TCA,
be worsening of neuropathic symptoms for the first few SNRI or an α-2-δ agonist should be considered for first-
weeks of application. Topical capsaicin (0.075%) applied line treatments, provided there are no contraindications.
sparingly three to four times per day to the affected area On the basis of trial data, duloxetine would be the
has been found to relieve neuropathic pain [49] (see preferred SNRI and pregabalin would be the preferred α-
above on use of lidocaine in painful DPN). 2-δ agonist. If pain is inadequately controlled, depending
upon contraindications, a different follow-up agent might
be considered as shown in Figure 1.
Pathogenetic treatments
Initial selection of treatment will be influenced
by the assessment of contraindications, consideration
Although several disease-modifying agents are under
of co-morbidities and cost; for example, in diabetic
investigation, only the anti-oxidant, α-lipoic acid, is
patients with a history of heart disease, elderly patients
supported by a meta-analysis and is available in certain
on other concomitant medications such diuretics and
countries [50]. Evidence supports the use of 600 mg
anti-hypertensives, patients with co-morbid orthostatic
i.v. per day over a 3-week period of this agent
hypotension and so on, TCA have relative contraindica-
in reducing neuropathic pain [50]. The meta-analysis
tions. In patients with liver disease, duloxetine should not
confirmed that the treatment was associated with a
be prescribed, and in those with oedema, pregabalin or
significant and clinically meaningful improvement in
gabapentin should be avoided (Table 3).
positive neuropathic symptoms as well as deficits. The
A combination of first-line therapies might be con-
results of long-term trials of oral α-lipoic acid for
neuropathic symptoms and deficits are eagerly awaited. sidered if there is pain, despite a change in first-line
monotherapy (Figure 1). If pain is inadequately con-
trolled, opioids such as tramadol and oxycodone might be
Previous guidelines on treatment added in a combination treatment.
of diabetic neuropathic pain
In a recent review on the management of neuropathic Non-pharmacological treatments
pain, Freynhagen and Bennett [51] provided treatment
recommendations based on recently published guidelines. A full review of studies on non-pharmacological treat-
They proposed a TCA (e.g. amitriptyline) or gabapentin ments for painful DPN is beyond the remit of this review

Copyright  2011 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2011; 27: 629–638.
DOI: 10.1002/dmrr
Painful Diabetic Peripheral Neuropathy 635

Painful diabetic neuropathy

Consideration of contraindications and comorbidities

α2-δ agonist SNRI


TCA
(pregabalin or gabapentin) (duloxetine)

If pain control is inadequate and considering contraindications

SNRI or α2-δ agonist TCA or α2-δ agonist


TCA or SNRI
(pregabalin or gabapentin) (pregabalin or gabapentin)

If pain control is still inadequate

Add opioid agonist as combination therapy


PDN = Peripheral diabetic neuropathy; SNRI = Serotonin-norepinephrine reuptake inhibitor;
TCA = Tricyclic antidepressants.

Figure 1. Treatment algorithm for painful diabetic peripheral neuropathy

Table 3. Tailoring treatment to the patient Bilateral sensory impairment is usually present. Other
cause of pain should be excluded and contraindications for
Factor Contraindication the use of individual drugs should be sought and catered
for. Based on trial evidence, this panel recommends that
Co-morbidities
Glaucoma TCAs
first-line therapies for painful DPN should be one of the
Orthostatic hypotension TCAs following:
Cardiovascular disease TCAs 1. a TCA
Hepatic disease Duloxetine
2. the SNRI duloxetine
Oedema Pregabalin, gabapentin
Unsteadiness and falls TCAs 3. the anti-convulsants pregabalin or gabapentin.
Other factors The decision should take into account patient co-
Cost Duloxetine, pregabalin morbidities and costs. Optimization of glycaemic control
Weight gain TCAs, pregabalin, gabapentin and aggressive management of cardiovascular risk factors
TCAs, tricyclic anti-depressants. are also clearly important. Combination therapy might
be useful for those with more severe pain, but there is
paucity of studies and further research is required. Studies
but suffice it to say that there are few well-designed tri-
are also required on direct head-to-head comparative
als in this area. However, lack of efficacy and unwanted
trials and long-term efficacy of drugs, as most trials
side effects from conventional drug treatments might
have lasted for less than 6 months. Key target areas
force many sufferers to try alternative therapies such
generating or modulating pain in painful DPN including
as acupuncture [55], near-infrared phototherapy [56],
peripheral small fibres with modulation at the level of
low-intensity laser therapy [57], transcutaneous electrical the spinal cord, the thalamus [22] and the other pain
stimulation [58], frequency-modulated electro-magnetic matrix areas in the brain require further studies in order
neural stimulation therapy [59], high-frequency exter- to develop more effective treatments. The association
nal muscle stimulation [60] and as a last resort, the of painful DPN with autonomic neuropathy also merits
implantation of an electrical spinal cord stimulator [61]. further investigation. Despite many trials looking for
effective pathogenetic treatments for painful DPN, only
the anti-oxidant intravenous α-lipoic acid has evidence
Conclusions from a meta-analysis and is in wide clinical use in
certain countries. Thus, further research is required to
Painful DPN is a significant clinical problem affecting up find novel and more effective pathogenetic treatments for
to a quarter of all diabetic patients and results in loss of painful DPN.
quality of life. Despite this, it continues to be under-
diagnosed and under-treated and this unsatisfactory
scenario must change. The minimum requirements for Conflict of interest
diagnosis of painful DPN are assessment of symptoms and
neurological examination, with shoes and socks removed. None declared.

Copyright  2011 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2011; 27: 629–638.
DOI: 10.1002/dmrr
636 S. Tesfaye et al.

Appendix Philip Low, MD, Department of Neurology, Mayo Clinic,


Rochester, MN, USA
The Toronto Consensus Panel on Diabetic Neuropathy Rayaz Malik, MD, Division of Cardiovascular Medicine,
James W Albers, MD, PhD, University of Michigan, Ann University of Manchester, Manchester, UK
Arbor, MI, USA Peter C. O’Brien, PhD, Mayo Clinic College of Medicine,
Gérard Amarenco, MD, Service de Rééducation Neu- Rochester, MN, USA
rologique et d’Explorations Périnéales, Hôpital Roth- Rodica Pop-Busui, MD, PhD, Department of Internal
schild, AP-HP, Paris, France Medicine, University of Michigan, Ann Arbor, MI, USA
Henning Anderson, MD, Department of Neurology, Bruce Perkins, MD, MPH, Division of Endocrinology, Uni-
Aarhus University Hospital, Aarhus, Denmark versity of Toronto, Toronto, ON, Canada
Joe Arezzo, PhD, Albert Einstein College of Medicine, Gerry Rayman, MD, Diabetes Centre, Ipswich Hospital,
New York, NY, USA Ipswich, UK
Misha-Miroslav Backonja, MD, Department of Neurology, James Russell, MD, Department of Neurology and Neuro-
University of Madison-Wisconsin, Madison, WI, USA physiology, University of Maryland, Baltimore, MD, USA
Luciano Bernardi, MD, Clinica Medica 1, Universita’ di Søren Sindrup, MD, Department of Neurology, Odense
Pavia, Pavia, Italy University Hospital, Odense, Denmark
Geert-Jan Biessels, MD, Department of Neurology, Rudolf Gordon Smith, MD, Department of Neurology, University
Magnus Institute, Utrecht, The Netherlands of Utah, Salt Lake City, UT, USA
Andrew J. M. Boulton, MD, Department of Medicine, Vincenza Spallone, MD, PhD, Department of Internal
University of Manchester, Manchester, UK Medicine, University of Tor Vergata, Rome, Italy
Vera Bril, MD, Department of Neurology, University of Martin Stevens, MD, Department of Medicine, University
Toronto, Toronto, ON, Canada of Birmingham, Birmingham, UK
Norman Cameron, PhD, University of Aberdeen, Solomon Tesfaye, MD, Diabetes Research Unit, Sheffield
Aberdeen, UK Teaching Hospitals, Sheffield, UK
Mary Cotter, PhD, University of Aberdeen, Aberdeen, UK Paul Valensi, MD, Service d’Endocrinologie-Diabétologie-
Peter J Dyck, MD, Department of Neurology, Mayo Clinic, Nutrition, Hôpital Jean Verdier, Bondy, France
Rochester MN, USA Tamás Várkonyi, MD, PhD, First Department of Medicine,
John England, MD, Department of Neurology at Louisiana
University of Szeged, Szeged, Hungary
State University Health Sciences Center, New Orleans,
Aristides Veves, MD, Beth Israel Deaconess Medical Cen-
LA, USA
ter, Harvard Medical School, Boston, MA, USA
Eva Feldman, MD, PhD, Department of Neurology, Uni-
Loretta Vileikyte, MD, PhD, Department of Medicine, Uni-
versity of Michigan, Ann Arbor, MI, USA
versity of Manchester, Manchester, UK
Roy Freeman, MD, Beth Israel Deaconess Medical Center,
Aaron Vinik, MD, PhD, Strelitz Diabetes Research Insti-
Harvard Medical School, Boston, MA, USA
tutes, Eastern Virginia Medical School, Norfolk, VA, USA
Simona Frontoni, MD, Department of Internal Medicine,
University of Tor Vergata, Rome, Italy Dan Ziegler, MD, Institute for Clinical Diabetology, Ger-
Jannik Hilsted, MD, Copenhagen University Hospital, man Diabetes Center at the Heinrich Heine University,
Copenhagen, Denmark Leibniz Center for Diabetes Research, Leibniz, Germany;
Michael Horowitz, MD, PhD, Department of Medicine, Department of Metabolic Diseases, University Hospital,
University of Adelaide, Adelaide, SA, Australia Düsseldorf, Germany
Peter Kempler, MD, PhD, I Department of Medicine, Sem- Doug Zochodne, MD, Department of Clinical Neuro-
melweis University, Budapest, Hungary science, University of Calgary, AB, Canada
Giuseppe Lauria, MD, Neuromuscular Diseases Unit, NIDDK observer – Teresa Jones, MD, NIDDK, Bethesda,
‘‘Carlo Besta’’ Neurological Institute, Milan, Italy MD, USA

References
1. Tesfaye S, Boulton AJM, Dyck PJ, et al. Complications Study Group. Relation- outcome period? Diabetes Care 2002;
Diabetic neuropathies: update on defini- ship between risk factors and mortality 25: 2010–2015.
tions, diagnostic criteria, estimation of in type 1 diabetic patients in Europe: 5. Tesfaye S, Boulton AJM (eds). Diabetic
severity and treatments. Diabetes Care the EURODIAB Prospective Complica- Neuropathy. Oxford: Oxford University
2010; 33: 2285–2293. Press: 2009.
tions Study (PCS). Diabetes Care 2008;
2. Boulton AJM, Malik RA, Arezzo J, 6. Treede R-D, Jensen TS, Campbell JN,
31(7): 1360–1366.
Sosenko JM. Diabetic somatic et al. Neuropathic pain: redefinition
4. Carrington AL, Shaw JE, Van
neuropathies. Diabetes Care 2004; 27: and a grading system for clinical and
1458–1486. Schie CMH, Abbott CA, Vileikyte L, research purposes. Neurology 2008; 70:
3. Soedamah-Muthu SS, Chaturvedi N, Boulton AJM. Can motor nerve 1630–1635.
Witte DR, Stevens LK, Porta M, conduction velocity predict foot prob- 7. Zelman DC, Brandenburg NA, Gore M.
Fuller JH; EURODIAB Prospective lems in diabetic subjects over a 6-year Sleep impairment in patients with

Copyright  2011 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2011; 27: 629–638.
DOI: 10.1002/dmrr
Painful Diabetic Peripheral Neuropathy 637

painful diabetic peripheral neuropathy. and small nerve fibers in diabetes. Dia- 37. Ray WA, Meredith S, Thapa PB, et al.
Clin J Pain 2006; 22: 681–685. betes Care 2006; 29: 883–887. Cyclic antidepressants and the risk of
8. Quattrini C, Tesfaye S. Understanding 22. Selvarajah D, Wilkinson ID, Gandhi R, sudden cardiac death. Clin Pharmacol
the impact of painful diabetic neu- Griffiths PD, Tesfaye S. Microvascular Ther 2004; 75: 234–241.
ropathy. Diabetes Metab Res Rev 2003; perfusion abnormalities of the Thala- 38. Kajdasz DK, Iyengar S, Desaiah D, et al.
19(Suppl 1): S2–S8. mus in painful but not painless diabetic Duloxetine for the management of
9. Vinik A, Zlateva G, Cheung R, Mur- polyneuropathy: a clue to the pathogen- diabetic peripheral neuropathic pain:
phy K, Emir B, Whalen E. Understand- esis of pain in type 1 diabetes. Diabetes evidence-based findings from post
ing the impact of pain response on Care 2011; 34(3): 718–20. hoc analysis of three multicentre,
changes in function, quality of life, 23. Gandhi R, Marques JLB, Selvarajah D, randomised, double-blind, placebo-
and sleep interference in patients with et al. Painful diabetic neuropathy is controlled, parallel-group studies. Clin
painful diabetic peripheral neuropathy associated with greater autonomic dys- Ther 2007; 29(Suppl 2): 536–546.
and post-herpetic neuralgia treated with function than painless diabetic neu- 39. Backonja MM, Beydoun A, Edwards KR,
pregabalin. J Pain 2010; 11: S17. ropathy. Diabetes Care 2010; 33: et al. Gabapentin for symptomatic treat-
10. Vileikyte L, Leventhal H, Gonzalez J, 1585–1590. ment of painful neuropathy in patients
et al. Diabetic peripheral neuropathy 24. Boulton AJM, Vinik AI, Arezzo JC, et al. with diabetes mellitus. JAMA 1998;
and depressive symptoms: the associ- Diabetes neuropathies: a statement by 280: 1831–1836.
ation revisited. Diabetes Care 2005; 28: the American Diabetes Association. Dia- 40. Freeman R, Durso-Decruz E, Emir B.
2378–2383. betes Care 2005; 28: 1–7. Efficacy, safety, and tolerability of
11. Vileikyte L, Peyrot M, Gonzalez JS, 25. Cruccu G, Sommer C, Anand P, et al. pregabalin treatment for painful dia-
et al. Predictors of depressive symp- EFNS guidelines on neuropathic pain betic peripheral neuropathy: findings
toms in persons with diabetic peripheral assessment: revised 2009. Eur J Neurol from seven randomised, controlled tri-
neuropathy: a longitudinal study. Dia- 2010; 17(8): 1010–1018. als across a range of doses. Diabetes Care
betologia 2009; 52: 1265–1273. 26. Bouhassira D, Attal N, Fermanian J, 2008; 31: 1448–1454.
12. Gore M, Brandenburg NA, Dukes E, et al. Development and validation of the 41. Donofrio PD, Raskin P, Rosenthal NR,
et al. Pain severity in diabetic peripheral neuropathic pain symptom inventory. et al.; CAPSS-141 Study Group. Safety
neuropathy is associated with patient Pain 2004; 108(3): 248–257. and effectiveness of topiramate for the
functioning, symptom levels of anxiety 27. Zelman DC, Gore M, Dukes E, Tai KS, management of painful diabetic periph-
and depression, and sleep. J Pain Symp- Brandenburg N. Validation of a modi-
eral neuropathy in an open-label exten-
tom Manage 2005; 30: 374–385. fied version of the brief pain inventory
sion study. Clin Ther 2005; 27(9):
13. Sankar A, Selvarajah D, Gandhi R, Ole- for painful diabetic peripheral neuropa-
1420–1431.
olo M, Emery C, Tesfaye S. Significant thy. J Pain Symptom Manage 2005;
42. Kastrup J, Petersen P, Dejard A, et al.
impact of mood disturbances of pain 29(4): 401–410.
Intravenous lidocaine infusion: a new
perception in painful DPN:time to re- 28. Backonja MM, Krause SJ. Neuropathic
treatment of chronic painful diabetic
evaluate current practice? Diabetologia pain questionnaire – short form. Clin J
neuropathy. Pain 1987; 28: 69–75.
2010; 53(Suppl 1): S446. Pain 2003; 19(5): 315.
43. Jarvis B, Coukell AJ. Mexilitene: a
14. Tesfaye S, Malik RA, Harris N, et al. 29. Bennett M. The LANSS pain scale:
review of its therapeutic use in painful
Arterio-venous shunting and proliferat- the leeds assessment of neuropathic
diabetic neuropathy. Drugs 1998; 56:
ing new vessels in acute painful neu- symptoms and signs. Pain 2001; 92:
691–708.
ropathy and rapid glycaemic control 147–157.
44. Harati Y, Gooch C, Swenson M, et al.
(insulin neuritis). Diabetologia 1996; 30. Melzack R. The short-form McGill pain
Double-blind randomised trial of tra-
39: 329–335. questionnaire. Pain 1987; 30: 191–197.
15. Vinik A. The approach to the manage- 31. Vileikyte L, Peyrot M, Bundy EC, et al. madol for the treatment of the pain of
ment of the patient with neuropathic The development and validation of a diabetic neuropathy. Neurology 1998;
pain. J Clin Endocrinol Metab 2010; neuropathy and foot ulcer specific Qual- 50: 1842–1846.
95(11): 4802–4811. ity of Life Instrument. Diabetes Care 45. Gimbel JS, Richards P, Portenoy RK.
16. Daousi C, McFarlane IA, Woodward A, 2003; 26: 2549–2555. Controlled-release oxycodone for pain
Nurmikko TJ, Bendred PE, Benbow SJ. 32. Vinik E, Hayes R, Oglesby A, Bastyr E, in diabetic neuropathy: a randomised
Chronic painful peripheral neuropathy Barlow P, Ford-Molvik S, Vinik A. The controlled trial. Neurology 2003; 60:
in an urban community: a control com- development and validation of the Nor- 927–934.
parison of people with and without dia- folk QOL-DN a new measure of patients’ 46. Bansal D, Bhansali A, Hota D,
betes. Diabet Med 2004; 21: 976–982. perception of the effects of diabetes and Chakrabarti A, Dutta P. Amitriptyline
17. Tesfaye S, Kempler P. Painful diabetic diabetic neuropathy. Diabetes Technol vs pregabalin in painful diabetic neu-
neuropathy. Diabetologia 2005; 48: Ther 2005; 7(3): 497–508. ropathy: a randomised double-blind
805–807. 33. Poole HM, Murphy P, Nurmikko TJ. clinical trial. Diabetic Med 2009; 26:
18. Oyibo SO, Prasad YDM, Jackson NJ Development and preliminary valida- 1019–1026.
Boulton AJM. The relationship between tion of the NePIQoL: a quality-of-life 47. Gilron I, Bailey JM, Tu D, et al. Nor-
blood glucose excursions and painful measure for neuropathic pain. J Pain triptyline and gabapentin, alone and
diabetic peripheral neuropathy: a pilot Symptom Manage 2009; 37: 233–245. in combination for neuropathic pain:
study. Diabet Med 2002; 19: 870–873. 34. Zigmond AS, Snaith RP. The hospital a double-blind, randomised controlled
19. Eaton SE, Harris ND, Ibrahim S, et al. anxiety depression scale. Acta Psychiatr crossover trial. Lancet 2009; 374(9697):
Differences insural nerve haemodynam- Scand 1983; 67: 361–370. 1252–1261.
ics in painful and painless neuropathy. 35. Finnerup NB, Sindrup SH, Jensen TS. 48. Gilron I, Bailey JM, Tu D, et al. Mor-
Diabetologia 2003; 934–939. The evidence for pharmacological treat- phine, gabapentin, or their combination
20. Quattrini C, Harris ND, Malik RA, Tes- ment of neuropathic pain. Pain 2010; for neuropathic pain. N Engl J Med 2005;
faye S. Impaired skin microvascular 150: 573–581. 352: 1324–1334.
reactivity in painful diabetic neuropa- 36. Sindrup S, Otto M, Finnerup NB, 49. Zhang WY, Li Wan PoA. The effective-
thy. Diabetes Care 2007; 30: 655–659. Jensen TS. Antidepressants in the treat- ness of topically applied capsaicin: a
21. Sorensen L, Molyneaux L, Yue DK. The ment of neuropathic pain. Basic Clin meta-analysis. Eur J Clin Pharm 1994;
relationship among pain, sensory loss, Pharmacol Toxicol 2005; 96: 399–409. 46: 517–522.

Copyright  2011 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2011; 27: 629–638.
DOI: 10.1002/dmrr
638 S. Tesfaye et al.

50. Ziegler D, Nowak H, Kempler P, pain in adults in non-specialist 58. Oyibo S, Breislin K, Boulton AJM. Elec-
Vargha P, Low PA. Treatment of symp- settings, March 2010. Available trical stimulation therapy through stock-
tomatic diabetic polyneuropathy with at: http://guidance.nice.org.uk/CG96 ing electrodes for painful diabetic
alpha-lipoic acid: a meta-analysis. Dia- [Accessed 26 July 2011]. neuropathy: a double-blind controlled
betic Med 2004; 21: 114–121. 55. Abuaisha BB, Constanzi JB, Boul- crossover study. Diabet Med 2004; 21:
51. Freynhagen R, Bennett MI. Diagnosis ton AJM. Acupuncture for the treatment 940–944.
and management of neuropathic pain. of chronic painful diabetic neuropathy: 59. Bosi E, Conti M, Vermigli C, et al. Effec-
BMJ 2009; 339: b3002. a long-term study. Diabetes Res Clin tiveness of frequency modulated elec-
52. Moulin DE, Clark AJ, Gilron I, et al. Pract 1998; 39: 115–121. tromagnetic neural stimulation in the
Pharmacological management of 56. Leonard DR, Farooqu MH, Myers S. treatment of diabetic peripheral neu-
chronic neuropathic pain – consensus Restoration of sensation, reduced pain, ropathy. Diabetologia 2005; 48(5):
statement and guidelines from the
and improved balance in subjects 817–823.
Canadian Pain Society. Pain Res Manage
with diabetic peripheral neuropathy: 60. Reichstein L, Labrenz S, Ziegler D, Mar-
2007; 12: 13–21.
a double-blind, randomised placebo- tin S. Effective treatment of symp-
53. Attal N, Cruccu G, Baron R, et al.; Euro-
controlled study with monochromatic tomatic diabetic polyneuropathy by
pean Federation of Neurological Soci-
infrared treatment. Diabetes Care 2004; high frequency external muscle stim-
eties. EFNS guidelines on the pharma-
cological treatment of neuropathic pain: 27: 168–172. ulation. Diabetologia 2005; 48(5):
2010 revision. Eur J Neurol 2010; 17(9): 57. Zinman LH, Ngo M, Ng ET, et al. Low- 824–828.
1113–e88. intensity laser therapy for painful 61. Tesfaye S, Watt J, Benbow SJ, et al.
54. NICE Clinical Guideline 96: Neu- symptoms of diabetic sensorimotor Electrical spinal-cord stimulation for
ropathic Pain. The pharmacolog- polyneuropathy∼: a controlled trial. painful diabetic peripheral neuropathy.
ical management of neuropathic Diabetes Care 2004; 27: 921–924. Lancet 1996; 348: 1698–1701.

Copyright  2011 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2011; 27: 629–638.
DOI: 10.1002/dmrr

You might also like