• 373
  • 0

 

 

 

Literature Review

Treatment of painful diabetic peripheral neuropathy

Casandra J Rosenberg1 and James C Watson2

Abstract

 

INTERNATIONAL
SOCIETY FOR PROSTHETICS AND ORTHOTICS

Prosthetics and Orthotics International

2015, Vol. 39(1) 17–28
© The International Society for Prosthetics and Orthotics 2014 Reprints and permissions: sagepub.co.uk/journalsPermissions.nav DOI: 10.1177/0309364614542266 poi.sagepub.com

 

 

page1image20160 page1image20320 page1image21080 page1image21504 page1image21664

Background: Painful diabetic peripheral neuropathy impairs quality of life and can be difficult to treat.
Objective: To discuss current treatment recommendations for painful diabetic peripheral neuropathy.
Study design: Literature review.
Methods: Systematic review of the literature discussing treatment of painful diabetic peripheral neuropathy. Existing treatment guidelines were studied and compared.

Results: Painful diabetic peripheral neuropathy occurs in about one in six people with diabetes. This condition impairs quality of life and increases healthcare costs. Treatment recommendations exist, but individual patient therapy can require a trial-and-error approach. Many treatment options have adjuvant benefits or side effects which should be considered prior to initiating therapy. Often, a combination of treatment modalities with various mechanisms of action is required for adequate pain control. Adequate medication titration and a reasonable trial period should be allowed. Conclusion: The treatment of painful diabetic peripheral neuropathy can be challenging, but effective management can improve patient’s quality of life.

Clinical relevance

Painful diabetic peripheral neuropathy impairs quality of life and can be difficult to treat. Many treatment options have adjuvant benefits or side effects which should be considered prior to initiating therapy. Often, a combination of treatment modalities with various mechanisms of action is required for adequate pain control.

Keywords

Diabetes, neuropathy, painful neuropathy, diabetic peripheral neuropathy Date received: 19 May 2014; accepted: 9 June 2014

 

 

Background

Diabetes affects more than 25 million people, in the United States representing greater than 11% of the adult popula- tion.1 Up to two-thirds of people with diabetes develop peripheral neuropathy, with an increasing prevalence with longer disease duration; 15%–25% of diabetic neuropa- thies are painful.2–6 Patients with painful neuropathies have healthcare costs three times greater than age matched controls.7 Diabetic peripheral neuropathy (DPN) and its sequelae, including ulceration and loss of limbs, cost the United States between 5 and 15 billion US dollars per year.8 Additionally, painful DPN significantly affects mul- tiple measures of quality of life and is highly associated with depression, anxiety, and sleep disturbances.2,9–11

Developing an individualized treatment plan for a per- son with painful DPN is challenging.12 Choosing a treat- ment can be difficult because of a lack of comparative

 

trials of effective treatments13,14 and limited or conflicting evidence for some treatments.13 Once a treatment is initi- ated, a patient can experience incomplete pain relief13,15 or suffer from the high side-effect profiles seen with many of the effective treatments.13,16

Over the past decade, the treatment of painful periph- eral neuropathies has evolved, and several treatment

1Department of Anesthesiology, Division of Pain Medicine, Mayo Clinic, Rochester, MN, USA

2Department of Neurology, Mayo Clinic, Rochester, MN, USA

Corresponding author:

Casandra J Rosenberg, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
Email: [email protected]

 

 

page1image46984

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

18

 

Prosthetics and Orthotics International 39(1)

 

 

page2image1352

algorithms have been established (Figure 1).16–20 Each of these guidelines considers treatment efficacy and adverse effects. The Canadian and the International Association for the Study of Pain Neuropathic Pain Special Interest Group (NeuPSIG) guidelines include ease of use and cost-effec- tiveness, while the NeuPSIG and the American Academy of Neurology (AAN) also consider quality-of-life out- comes. One of the guidelines is strictly for painful DPN,20 and three others include specific subgroup recommenda- tions for DPN.16,18,19

Disease management

The first step in the management of painful DPN is treat- ing the cause of neuropathy, the diabetes. There is evi- dence that tight glucose control, particularly in the early stages of diabetes, can delay progression of painful DPN.21 A recent Cochrane review showed a relative risk of devel- oping DPN of 0.35 for patients who undergo intensive glu- cose control compared to conventional treatment (95% confidence interval (CI) = 0.23–0.53; p < 0.00001; I2 = 0%; 1203 participants; three trials).22 Neuropathic pain symp- toms may improve with tight glycemic control; however, if this occurs, the reversal of symptoms is limited and the primary goal of disease treatment is to prevent or slow pro- gression. Comorbid conditions such as depression, anxi- ety, sleep impairment, and renal dysfunction can affect pain and should also be addressed.11,13,23,24

Medications

Careful consideration of several patient factors can improve success in medical therapy of painful DPN. First, the patient’s medical comorbidities should be considered. Limiting factors such as pulmonary, cardiovascular, renal, or hepatic impairment should be identified as these may influence medication choice. In certain patients, physio- logic testing (such as electrocardiogram (EKG) prior to initiating tricyclic antidepressants (TCAs)) or laboratory testing (such as creatinine or liver function tests) may be required prior to medication initiation. Next, comorbid conditions which might benefit from the same medication used to treat the painful DPN, such as insomnia, depres- sion, fibromyalgia, or headache, should be noted as treat- ing two processes with one medication would be preferable to initiating separate treatments. Finally, the medication side-effect profile should be considered, and the risk-to- benefit ratio weighed.

Neuropathic pain medications rarely resolve pain. However, even a modest to moderate decrease in the patient’s reported pain level can be clinically meaningful and improve a patient’s quality of life. An improvement of 30% or 2 points on a 0–10 Likert scale has been found to be clinically meaningful to patients.25 The practitioner and the patient should discuss expectations and treatment goals to maximize outcomes. When a neuropathic pain medica- tion is tolerated and partially effective, but insufficient to

 

improve function, a second agent with a distinct mecha- nism of action should be added. Many studies show an additive benefit when combining medications from differ- ent classes.26–28 Most treatment guidelines recommend adding an additional agent rather than transitioning from one agent to another, particularly if some benefit was achieved from the initial medication.18,19

TCAs

Mechanism. TCAs prevent the reuptake of norepinephrine and serotonin; antagonize N-methyl-d-aspartate (NMDA), 5-HT, histamine, muscarinic, and alpha-adrenergic recep- tors; and inhibit sodium and l-type calcium channels.16 Each of these effects may have an impact on pain regulation.

Additional benefits. TCAs are approved for the treatment of depression by the US Food and Drug Administration (FDA), though dosing needed for therapeutic benefit in painful DPN is usually lower than that required for the treatment of depression.29 Additional off-label uses include insomnia, chronic myofascial pain, fibromyalgia, migraine headache, irritable bowel syndrome, and smoking cessation.30–32

Side effects. The most common TCA side effects are dry mouth, decreased gastrointestinal (GI) motility, orthostatic hypotension, weight gain, urinary retention, and cognitive dysfunction.33 Sedation is common and nighttime dosing may improve sleep and limit daytime drowsiness. Toler- ance to side effects develops, and a low starting dose with gradual up-titration is recommended.19 The most severe side effects are cardiac dysrhythmias, and an EKG to screen for conduction system disease in elderly or at-risk patients is recommended prior to initiation of therapy.34,35 As with other antidepressants, there is a slightly increased risk of suicidal thoughts or actions in young adults.36 TCAs confer a risk of serotonin syndrome when used with other medications that affect serotonin (other antidepressants, tramadol, tapentadol, some antiemetics) and should be used with caution at lowered goal dosages when used in combination with other serotonergic agents. TCAs can also lower the seizure threshold.37

DPN evidence. All of the medication treatment guidelines include the TCAs as first-line medications unless contrain- dicated, with one exception.20 TCAs have robust evidence supporting analgesic efficacy in painful neuropathies com- pared to placebo.12 Head-to-head trials have shown equal analgesic efficacy to gabapentin19,26,38

Serotonin–norepinephrine reuptake inhibitors

Mechanism. Serotonin–norepinephrine reuptake inhibitors (SNRIs) affect the balance of these central nervous system neurotransmitters, which is thought to promote

 

 

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

Rosenberg

 

19

 

 

page3image1200

page3image1880 page3image2304 page3image2464 page3image2888 page3image3048 page3image3472 page3image3896 page3image4320 page3image4480 page3image4904 page3image5664 page3image6256 page3image6680

 

Mild

Topical agents Oral supplements Therapeuc modalies

 

Symptoms persist:

Side effects prohibit treatment:

 

Confirm diagnosis

Evaluate for treatable causes of neuropathy & opmize disease management

Consider comorbid condions:

Depression / Anxiety Sleep dysfuncon Fibromyalgia Headache

Symptom severity:

Moderate

Use first line neuropathic pain adjuvant:

Gabapenn Pregabalin
TCA
Duloxene

and consider mild treatment

agents

Symptoms persist despite adequate dose and trial period:

Add another first line neuropathic pain adjuvant with different mechanism of acon

Symptoms persist despite adequate dose and trial period:

Consider pain consultaon & advanced intervenons

 

Severe, necessitang immediate analgesia

1 moderate treatment agent, plus:

Tramadol/Tapentadol

If above contraindicated:

Opioid

Symptoms improve:

Taper analgesic / opioid if possible

 

 

page3image19152 page3image19312 page3image19736 page3image19896 page3image20320 page3image20744 page3image21168 page3image21592 page3image22016 page3image22608 page3image23032 page3image23456 page3image23880 page3image24304

Try new agent

 

 

page3image25256 page3image26016

Figure 1. Treatment algorithm for painful diabetic peripheral. TCA: tricyclic antidepressant.

anti-nociception and reduce pro-nociception.39 While the primary mechanism of action is similar to TCAs, dosing is generally less limited by anticholinergic, antiadrenergic, or muscarinic side effects.

 

Additional benefits. Duloxetine is a balanced SNRI (block- ing serotonin and norepinephrine equally) that is approved by the FDA for painful DPN, fibromyalgia, chronic mus- culoskeletal pain, depression, and anxiety.40,41 A potential

 

 

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

20

 

Prosthetics and Orthotics International 39(1)

 

 

page4image1352

advantage of duloxetine is that the therapeutic dosages are similar for each condition such that it can be used to treat two comorbid conditions without dosage adjustment. Other SNRIs, including venlafaxine, may have similar neuropathic pain treatment effects, but have not been as rigorously studied and are only FDA approved for depres- sion and anxiety.

Side effects. Common SNRI side effects include GI upset, headache, and insomnia. Unique side effects with duloxe- tine are hyperhidrosis and a possible increase in bleeding risk.42,43 Side effects are typically less bothersome than those seen with TCAs.41 As with other antidepressants, there is a slightly increased risk of suicidal thoughts or actions in young adults.36 Like TCAs, SNRIs also decrease the seizure threshold, and their serotonergic effect gives a risk of serotonin syndrome.44

Evidence. Two of the above guidelines list SNRIs as first- line treatment options.16,18 Studies have suggested similar efficacy of duloxetine to amitriptyline, with a slightly bet- ter side-effect profile.45

Calcium channel blockers: gabapentin and pregabalin

Mechanism. Gabapentin and pregabalin block the presyn- aptic calcium channel at the α2δ ligand, inhibiting release of excitatory neurotransmitters. Additionally, they may antagonize NMDA receptors associated with pain signaling.46,47

Additional benefits. Pregabalin is FDA approved for the treatment of painful DPN, post-herpetic neuralgia, central neuropathic pain, and fibromyalgia.48 Gabapentin is FDA approved for post-herpetic neuralgia but is also commonly used for other neuropathic pain states.49 Other benefits of the gabapentanoids are improved sleep50 and possibly anxiolysis.51

Side effects. Common gabapentin and pregabalin side effects include cognitive slowing, drowsiness, dizziness, and peripheral edema.17 Pregabalin can cause euphoria in a very small percentage of patients, which may be a con- sideration in patients with psychiatric comorbidities.52 As both gabapentin and pregabalin are renally excreted, dos- age adjustment is required in the setting of renal impair- ment. There are minimal significant medication interactions. As with antidepressants and other antiepilep- tic medications, there is a slightly increased risk of suicidal thoughts or actions in young adults.49

Evidence. Gabapentin has been shown to be as efficacious as the TCA nortriptyline.26 Gabapentin or pregabalin are recommended as first-line medications in the majority of

 

the above guidelines17–20 with one guideline giving prefer- ence to pregabalin over gabapentin or antidepressants,20 and another as a second choice if TCAs are contraindi- cated.16 One guideline recommends earlier-generation anticonvulsants first; however, gabapentin and pregaba- lin’s minimal drug interactions and reduced requirement for monitoring can be advantages over other anticonvulsants.53

Opioids, including tapentadol and tramadol

Mechanism. Opioid medications bind to μ and other opioid receptors on neuronal cell membranes, modulating descending inhibitory and ascending excitatory pain mechanisms. Their primary analgesic targets are in the spi- nal cord and elsewhere in the central nervous system, but receptors throughout the body are responsible for analge- sia and opioid-related side effects.

Tapentadol is a partial μ agonist with a norepinephrine reuptake inhibitory effect.54 Tramadol is also a partial μ receptor agonist and has both serotonin and norepineph- rine reuptake inhibition.55,56

Additional benefits. The main therapeutic benefit of opioid medications is analgesia. Opioid medications should not be used for insomnia, anxiolysis, or other indications.

Tapentadol is FDA approved for painful DPN.57 It is postulated that the combined μ receptor activation and norepinephrine reuptake inhibition of tramadol and tapen- tadol offers additional benefit compared to the single- mechanism agents.58,59

Side effects. A primary concern in the prescription of opi- oid medications is their potential for misuse. Opioids acti- vate the brain’s mesolimbic reward center, resulting in feelings of pleasure which can lead to craving and compul- sive use.60 Opioids are the most commonly misused pre- scription medication in the United States and are responsible for the majority of prescription drug–related acute hospital admissions and drug treatment center admis- sions.61 Tolerance and physical dependence, resulting in the need for gradually increasing dosage as well as with- drawal symptoms if the medication is decreased or discon- tinued, are an inherent physiologic response to the medication and should not be confused with addiction. Addiction involves craving and compulsive use despite harm.62

The most serious side effect of opioid medications is respiratory depression. The effect can be additive with other substances such as benzodiazepines and alcohol. Other side effects include nausea, vomiting, pruritus, con- stipation, sedation, and dizziness. Overdose or accumula- tion of breakdown products can produce myoclonus and contribute to sedation and respiratory depression. Over time, immune and endocrine dysfunction, including

 

 

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

Rosenberg

 

21

 

 

page5image1192

hypotestosteronism, occur. Another potential issue with chronic opioid use is opioid-induced hyperalgesia, a state of increased sensitivity to pain and worsening of pain while receiving opioids. This can be particularly problem- atic in patients who already struggle with pain issues.63 Opioids should be used with caution in renal and hepatic impairment, particularly with extended or continuous release formulations.64,65

Tramadol and tapentadol can contribute to serotonin syndrome and can lower the seizure threshold. They should also be used with caution in patients with renal and hepatic impairment and in those receiving other medications with serotonergic effects.66–68

Evidence. There is evidence that opioid medications are effective in painful DPN,69–71 including tramadol55,56 and tapentadol.72 Notably, most studies were carried out with extended or continuous release formulations.55,69,70,72 Opi- oids are included in the treatment algorithms as later-tier choices due to issues with long-term use and risks.20 Their later-tiered position in the treatment algorithms is large part related to the potential adverse effects and chronic use risks inherent with opioids. However, they may be useful in appropriately selected and monitored patients. In a crossover trial for post-herpetic neuralgia, patients pre- ferred extended release morphine to nortriptyline.73

There are multiple established guidelines for opioid pre- scribing risk mitigation.74 Similar to other painful DPN treatments, an opioid trial should include pre-determined goals for pain and functional improvement. A treatment agreement should be agreed upon and maintained, and reg- ular screening for aberrant use, abuse, misuse, and diver- sion should be completed, including urine drug screening to confirm appropriate medication and breakdown products and to screen for other medication or substance use that could increase the patient’s treatment-related risk.74

Topical agents

Topical agents carry the advantage of limited systemic side effects, making them excellent first-line or adjuvant pain medications. Topical creams and gels can cover a wide area. Patches can be less practical given their limited size and the typical distribution of DPN involving the feet.

Topical capsaicin, including high-dose patches (Qutenza)

Mechanism. Capsaicin is an active component of chili pep- pers. It binds to the TRPV1 heat-activated calcium channel and causes it to open at normal body temperature. Pro- longed activation of the receptor results in the depletion of neurotransmitters, particularly substance P, and reduces the ability of neurons to transmit pain signals. Standard capsai- cin creams come in concentrations of 0.025%–0.25%.

 

They must be applied multiple times per day for several weeks before analgesic effects become significant. Qutenza is a capsaicin patch with an 8% concentration. It is applied after appropriate local analgesia with topical lidocaine and left in place for 60 minutes. A single applica- tion of the Qutenza patch may provide up to 3 months of pain relief.

Side effects. Skin irritation is the most common side effect and many patients do not tolerate capsaicin treatment.75,76 During and after administration of the 8% capsaicin patch, pain can be significant (like a severe sunburn) and may require medical management with opioids for a number of days following application.77 Extreme caution must be used to avoid contact of any concentration of capsaicin with the eyes or mucous membranes.

Evidence. A meta-analysis of studies reported a pooled 2.35 odds ratio in favor of capsaicin over placebo (95% CI = 1.48–3.22).12 High-dose capsaicin was most effective in patients with post-herpetic and human immunodefi- ciency virus–related neuropathies.78 Qutenza is FDA approved for post-herpetic neuralgia.

Lidocaine, including patches

Mechanism. Lidocaine blocks voltage-gated sodium chan- nels, which stops propagation of action potentials in neu- rons.79 A 5% concentration patch is available.

Side effects. With 12 h of continuous wear of the 5% patch, only 3% of the total dose is absorbed, making systemic side effects unlikely.80 Skin irritation is the most common side effect.80

Evidence. The lidocaine 5% patch is FDA approved for post-herpetic neuralgia. The patch has been found to reduce pain and improve quality of life in DPN.81 Lido- caine patches were found to give a similar degree of ben- efit as pregabalin to patients with DPN, as well as additional benefit when used in conjunction with pregabalin.82

Other topical agents

Menthol. Menthol transiently activates TRPM8 ion chan- nels, causing an endogenous cooling-induced analgesia.83 There are no randomized controlled trials for neuropathic pain; case reports and small studies have shown some benefits.84,85

Amitriptyline, ketamine and baclofen. These medications are typically compounded in a cream or pluronic lecithin organogel base for topical application. Studies show no systemic toxicities, but have mixed results, at best, for the treatment of neuropathic pain.86–89

 

 

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

22

 

Prosthetics and Orthotics International 39(1)

 

 

page6image1456 page6image1616 page6image1776 page6image1936 page6image2096 page6image2256 page6image2416 page6image2576 page6image2736 page6image2896 page6image3056 page6image3216 page6image3376 page6image3536

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

 

Table 1. Prescribing recommendations for medications for painful diabetic peripheral neuropathy.

 

 

Agent

 

Dosage and titration recommendations

 

Maximum dosage

 

Most common side effects

 

Precautions

 

Other benefits

 

 

Amitriptyline/ nortriptyline

 

10–25 mg at bedtime, increase every 4–7 days to goal of 100 mg at bedtime

 

150 mg daily

 

Sedation, dry mouth, orthostatic hypotension, urinary retention, weight gain, confusion, blurred vision, thrombocytopenia

 

Small risk of emerging suicidality (boxed warning), caution in patients with cardiac disease (high dosages >100 mg daily associated with sudden cardiac death and dysrhythmias), serotonin syndrome, glaucoma

 

Improvement of depression (though typical doses for pain are lower than doses necessary for depression)

 

 

Duloxetine

 

30 mg once daily for one week, then increase to 60 mg in single or divided doses

 

60mg twice daily

 

Fatigue, nausea, hyperhidrosis

 

Small risk of emerging suicidality (boxed warning), increased bleeding risk (care with anticoagulants), withdrawal syndromes with abrupt discontinuation, serotonin syndrome, hepatic failure

 

Treats depression and anxiety (at similar dosage used for pain), as well as myofascial pain

 

 

Gabapentin

 

300mg at bedtime, increase every
4–7 days by 300mg increments initially to three times per day dosing, then to goal of 1800mg/day, as necessary to 3600 mg/day
50mg three times daily or 75mg twice daily, after first 4–7 days increase
by same to goal of 300 mg/day, as necessary to 600mg/day
50mg twice daily; increase every
4–7 days to a maximum of 100 mg per dose four times per day

 

3600 mg/day (divided into three doses)

 

Sedation, confusion, dizziness, peripheral edema, tremor

 

Renal insufficiency (must dose adjust), withdrawal syndromes/seizure with abrupt discontinuation

 

Improvement of sleep disturbance, anxiety, hot flashes; no clinically significant drug interactions

 

 

Pregabalin

 

600 mg/day

 

Sedation, confusion, dizziness, peripheral edema, euphoria

 

Renal insufficiency (must dose adjust), psychiatric disease (euphoria risk), withdrawal syndromes/seizure with abrupt discontinuation

 

Improvement of sleep disturbance, fibromyalgia, and anxiety; no clinically significant drug interactions
Rapid onset analgesic benefit

 

 

Tramadol

 

400 mg/day

 

Nausea/vomiting, constipation, dizziness, seizures at dosages >400mg/day

 

History of substance abuse, sedation, driving impairment, seizure disorder, risk of serotonin syndrome with concomitant use of SSRI, SNRI, and TCAs

 

 

Tapentadol

 

50mg every 4h as needed; increase every 4–7 days to a maximum of 100mg every 4h as needed

 

600mg/day

 

Nausea/vomiting, constipation, dizziness, headache, seizure, hypotension

 

History of substance abuse, sedation, respiratory depression, driving impairment, seizure disorder,
risk of serotonin syndrome with concomitant use of SSRI, SNRI, and TCAs; hepatic dysfunction

 

Rapid onset analgesic benefit

 

 

Opioids (morphine, oxycodone)

 

15 mg oral immediate release morphine (or another opioid of equianalgesic dose such as 10mg oxycodone) three to four times per day; after 1–2 weeks convert total daily dose to long acting formulation. Continue short acting as needed

 

No maximum dosage

 

Constipation, nausea/ vomiting, drowsiness, confusion, dizziness, pruritus

 

History of substance abuse, driving impairment, tolerance, abuse, diversion

 

Rapid onset analgesic benefit

 

 

Alpha-lipoic acid

 

600 mg once daily

 

Nausea, rash, and hypothyroidism

 

Hypoglycemia

 

Few toxicities

 

 

 

Rosenberg

 

23

 

 

page7image1304

Other supplements

Alpha-lipoic acid. A lipophilic antioxidant, alpha-lipoic acid (ALA), has been shown to improve nerve blood flow and distal conduction and to reduce oxidative stress.90 The pri- mary side effect is nausea. Both oral (at a dosage of 600 mg per day) and intravenous supplementation have been shown to improve symptoms of painful DPN.91,92

Acetyl-l-carnitine. Acetyl-l-carnitine (ALC) has shown improved peripheral nerve conduction velocity and nerve regeneration, thought to be due to its antioxidant properties and role in catabolism/anabolism.93 Several studies have shown benefit in patients with painful DPN (at a dosage of 1000 mg three times daily by mouth) (Table 1).94–96

Therapeutic modalities

Acupuncture

Acupuncture has been shown to be effective for painful DPN in small trials without placebo control.97–99

Transcutaneous electrical nerve stimulation

Transcutaneous electrical nerve stimulation (TENS) deliv- ers an electrical current via electrodes on the skin arranged about the affected area. The intensity, frequency, and pat- tern of stimulation can be adjusted by the patient or thera- pist. Several studies including sham treatment arms have demonstrated benefit in painful DPN.100–102

Cognitive behavioral therapy

Cognitive behavioral therapy (CBT) replaces dysfunc- tional thoughts and behaviors with positive ones and has been found effective in multiple pain states.103–105 One ran- domized pilot study showed benefit for patients with pain- ful DPN as compared to typical management.106

Advanced interventions

In addition to medications, several interventions are gaining interest in the treatment of painful neuropathies. Interventional treatments have previously been described as “invasive procedures involving delivery of drugs into tar- geted areas, or ablation/modulation of targeted nerves.”13 Recently, the NeuPSIG compiled evidence for interven- tional treatments in the management of neuropathic pain, including a subgroup analysis for painful DPN in combina- tion with other painful peripheral neuropathies.

Neuromodulation

Neuromodulation involves the application of electrical cur- rent to neural structures to modulate neural transmission.

 

 

page7image19816 page7image19976 page7image20136 page7image20296 page7image20456 page7image20616 page7image20776 page7image20936 page7image21096 page7image21256 page7image21416 page7image21576 page7image21736

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

 

Table 1. (Continued)

 

 

Agent

 

Dosage and titration recommendations

 

Maximum dosage

 

Most common side effects

 

Precautions

 

Other benefits

 

 

Acetyl-l-carnitine Topical

 

1000 mg three times per day

 

Agitation, nausea, vomiting, bloating
Local skin problems such as erythema, burning, or pain (patients frequently require prescription analgesics for 1–2 weeks after application)

 

Few toxicities
No known systemic toxicities

 

 

capsaicin (high concentration 8% patch)

 

Before patch application, use topical anesthetic for 60 min, remove, and confirm anesthesia. Place patch over symptomatic area for 60 min. Should be placed by medical staff trained in its usage using non-latex gloves. May cut patch to shape. Following treatment, wash with provided soap.

 

May apply up to four patches at one time.

 

Unknown risk from repeated exposures, not recommended for face or over broken skin

 

 

Topical capsaicin (0.075%–0.75% cream)
Topical lidocaine (5% patch)

 

Apply cream to affected areas four times daily

 

Local skin problems such as erythema, burning, or pain
Local skin problems such as burning, irritation

 

Not recommended for face or around mucous membranes or over broken skin; apply wearing gloves Not recommended around mucous membranes or over broken skin

 

No known systemic toxicities Low risk of systemic toxicity

 

 

Apply up to three patches for 12 h daily; may cut to shape

 

 

SSRI: selective serotonin reuptake inhibitor; SNRI: serotonin and norepinephrine reuptake inhibitors; TCA: tricyclic antidepressant. Based on information from Dworkin et al.18.

 

 

 

24

 

Prosthetics and Orthotics International 39(1)

 

 

page8image1352

The most common method of neuromodulation for pain is dorsal column stimulation, also known as spinal cord stimu- lation (SCS). Small electrodes are implanted in the epidural space and connected to a battery-operated pulse generator implanted in the flank or abdominal wall. Prior to implanta- tion, a trial of stimulation with temporary percutaneous electrodes is undertaken to ensure effective pain relief. Several non-randomized case series have demonstrated pain relief of peripheral neuropathy even in medically resistant cases.107–109 A single study with a crossover-type design showed benefit compared to sham stimulation.108

For isolated mononeuropathies, placement of a tradi- tional stimulator lead immediately adjacent to the affected nerve has been described.110

Intrathecal drug delivery systems

Intrathecal drug delivery systems (IDDS) use a surgically implanted pump attached to a small, flexible catheter tun- nelled into the cerebrospinal fluid (CSF). Morphine and ziconotide (a selective N-type voltage-gated calcium chan- nel blocker) are FDA-approved medications for intrathecal infusion.111,112

There are currently no studies showing evidence for intrathecal drug delivery specifically for neuropathic non- cancer pain. Given concentrated drug delivery into the CSF, intrathecal systems may be useful when systemic side effects from oral analgesics or adjuvants are limiting.

Factors that increase the risk of IDDS implantation in diabetic patients include impaired wound healing, relative immunosuppression increasing the risk of infection, and comorbid cardiovascular or renal disease. If an intrathecal drug delivery system is being considered in a diabetic patient, it is recommended that their glucose control is optimized prior to implantation.113 Patients with an intrath- ecal system need to be able to maintain regular follow-up for system refills and adjustments.

Conclusion

While painful DPN can prove difficult to treat, there are a variety of therapeutic options, including behavioral thera- pies, physical modalities, topical and oral medications, and implantable devices. There are several key concepts for developing a treatment approach.

Tight glycemic control is important to delay onset and progression of painful DPN. Consideration of comorbid conditions including sleep disturbance, depression, and other pain states is important both to minimize the pain of DPN itself and when selecting the most appropriate medi- cal therapy. Many painful DPN treatments can also address other comorbid conditions. Furthermore, some medication side effects could exacerbate preexisting problems and should be avoided.

 

Managing expectations is vital when starting or chang- ing medications. Most treatments are considered success- ful if they provide 30%–50% improvement or at least 2 points on a 10-point Likert scale. For some patients, this improvement will be worthwhile, while others may weigh the benefits less favorably relative to the side effects of the medication.

Medication side effects can be a barrier to treatment success. A frank conversation discussing common or seri- ous side effects should be had prior to starting a new treat- ment. Many of these treatment side effects can be improved over time due to progressive tolerance; these can be miti- gated at the start of treatment by gradual up-titration of the medication dose. Each medication should be maintained at the lowest effective dose.

For patients who have had prior medication trials to treat their painful DPN, it is important to ensure that their trial was adequate in dosage achieved and duration of treatment (several weeks at goal dosage). It is common that a prior trial was inadequate and that a retrial may be appropriate.

Finally, an important strategy to the management of painful DPN is multimodal therapy. Many studies show benefit from combination therapies. This strategy is likely effective due to the additive effects of pain relief from multiple mechanisms of action. It is important to carefully consider the individual treatments to avoid unpleasant or life-threatening side effects, such as serotonin syndrome with combinations of high-dose TCAs and tramadol. Topical medications, behavioral strategies, and physical modalities can be particularly beneficial in combination with other treatments due to their negligible side-effect profiles.

Author contribution

All authors contributed equally in the preparation of this manuscript.

Declaration of conflicting interests

None declared.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

References

  1. Centers for Disease Control and Prevention. National diabe- tes facts sheet, national estimates and general information on diabetes and pre-diabetes in the United States. Atlanta, GA: US Department of Health and Human Services, Centers for Disease Control and Prevention, 2011.

  2. Argoff CE, Cole BE, Fishbain DA, et al. Diabetic peripheral neuropathic pain: clinical and quality-of-life issues. Mayo Clin Proc 2006; 81: S3–S11.

 

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

Rosenberg 25

 

 

page9image896

  1. Partanen J, Niskanen L, Lehtinen J, et al. Natural history of peripheral neuropathy in patients with non-insulin-dependent diabetes mellitus. N Engl J Med 1955; 333: 89–94.

  2. Dyck PJ, Kratz KM, Karnes JL, et al. The prevalence by staged severity of various types of diabetic neuropathy, retinopathy, and nephropathy in a population-based cohort: the Rochester Diabetic Neuropathy Study. Neurology 1993; 43: 817–824.

  3. Gregg EW, Sorlie P, Paulose-Ram R, et al. Prevalence of lower-extremity disease in the US adult population >=40 years of age with and without diabetes: 1999–2000 national health and nutrition examination survey. Diabetes Care 2004; 27: 1591–1597.

  4. Van Acker K, Bouhassira D, De Bacquer D, et al. Prevalence and impact on quality of life of peripheral neuropathy with or without neuropathic pain in type 1 and type 2 diabetic patients attending hospital outpatients clinics. Diabetes Metab 2009; 35: 206–213.

  5. Berger A, Dukes EM and Oster G. Clinical characteristics and economic costs of patients with painful neuropathic dis- orders. J Pain 2004; 5: 143–149.

  6. Gordois A, Scuffham P, Shearer A, et al. The health care costs of diabetic peripheral neuropathy in the US. Diabetes Care 2003; 26: 1790–1795.

and guidelines from the Canadian Pain Society. Pain Res

Manag 2007; 12: 13–21.
20. Bril V, England J, Franklin GM, et al. Evidence-based

guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Neurology 2011; 76: 1758–1765.

21. The effect of intensive diabetes therapy on the development and progression of neuropathy. The Diabetes Control and Complications Trial Research Group. Ann Intern Med 1995; 122: 561–568.

22. Fullerton B, Jeitler K, Seitz M, et al. Intensive glucose con- trol versus conventional glucose control for type 1 diabetes mellitus. Cochrane Database Syst Rev 2014; 2: CD009122.

23. Bair MJ, Robinson RL, Katon W, et al. Depression and pain comorbidity: a literature review. Arch Intern Med 2003; 163: 2433–2445.

24. Santoro D, Satta E, Messina S, et al. Pain in end-stage renal disease: a frequent and neglected clinical problem. Clin Nephrol 2013; 79(Suppl. 1): S2–S11.

25. Farrar JT, Young JP Jr, LaMoreaux L, et al. Clinical impor- tance of changes in chronic pain intensity measured on an 11-point numerical pain rating scale. Pain 2001; 94: 149–

 

 

9. Benbow SJ, Wallymahmed ME and MacFarlane IA. 158.

 

 

Diabetic peripheral neuropathy and quality of life. QJM

1998; 91: 733–737.

  1. Galer BS, Gianas A and Jensen MP. Painful diabetic poly-

    neuropathy: epidemiology, pain description, and quality of

    life. Diabetes Res Clin Pract 2000; 47: 123–128.

  2. Sadosky A, Schaefer C, Mann R, et al. Burden of illness associated with painful diabetic peripheral neuropathy among adults seeking treatment in the US: results from a retrospective chart review and cross-sectional survey.

    Diabetes Metab Syndr Obes 2013; 6: 79–92.

  3. Kingery WS. A critical review of controlled clinical trials for peripheral neuropathic pain and complex regional pain

    syndromes. Pain 1997; 73: 123–139.

  4. Dworkin RH. Inclusion criteria, http://www.acc.co.nz/

    for-providers/clinical-best-practice/interventional-pain- management/interventions/intervention-index/ WCM1_034233 (2013, accessed 27 March 2014).

  5. O’Connor AB and Dworkin RH. Treatment of neuropathic pain: an overview of recent guidelines. Am J Med 2009; 122: S22–S32.

  6. Davies HT, Crombie IK, Lonsdale M, et al. Consensus and contention in the treatment of chronic nerve-damage pain. Pain 1991; 47: 191–196.

  7. Finnerup NB, Otto M, McQuay HJ, et al. Algorithm for neu- ropathic pain treatment: an evidence based proposal. Pain 2005; 118: 289–305.

  8. Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revi- sion. Eur J Neurol 2010; 17: 1113–e88.

  9. Dworkin RH, O’Connor AB, Audette J, et al. Recommendations for the pharmacological management of neuropathic pain: an overview and literature update. Mayo Clin Proc 2010; 85: S3–S14.

  10. Moulin DE, Clark AJ, Gilron I, et al. Pharmacological man- agement of chronic neuropathic pain—consensus statement

26. Gilron I, Bailey JM, Tu D, et al. Nortriptyline and gabap- entin, alone and in combination for neuropathic pain: a double-blind, randomised controlled crossover trial. Lancet 2009; 374: 1252–1261.

27. Simpson DA. Gabapentin and venlafaxine for the treatment of painful diabetic neuropathy. J Clin Neuromuscul Dis 2001; 3: 53–62.

28. Gilron I, Bailey JM, Tu D, et al. Morphine, gabapentin, or their combination for neuropathic pain. N Engl J Med 2005; 352: 1324–1334.

29. Furukawa TA, McGuire H and Barbui C. Meta-analysis of effects and side effects of low dosage tricyclic antidepres- sants in depression: systematic review. BMJ 2002; 325: 991.

30. Prochazka AV, Kick S, Steinbrunn C, et al. A randomized trial of nortriptyline combined with transdermal nicotine for smoking cessation. Arch Intern Med 2004; 164: 2229–2233.

31. Micromedex 2.0 (Amitriptyline Hydrochloride) Available at: http://www.micromedexsolutions.com/micromedex2/ librarian/ND_T/evidencexpert/ND_PR/evidencex- pert/CS/54AC7A/ND_AppProduct/evidencexpert/ DUPLICA TIONSHIELDSYNC/AEFE69/ND_PG/evi- dencexpert/ND_B/evidencexpert/ND_P/evidencexpert/ PFActionId/evidencexpert.DoIntegratedSearch?Search Term=amitriptyline (accessed 30 April 2014).

32. Micromedex 2.0 (Nortriptyline Hydrochloride) Available at: http://www.micromedexsolutions.com/micromedex2/ librarian/ND_T/evidencexpert/ND_PR/evidencex- pert/CS/B42175/ND_AppProduct/evidencexpert/ DUPLICA TIONSHIELDSYNC/56E6AD/ND_PG/evi- dencexpert/ND_B/evidencexpert/ND_P/evidencexpert/ PFActionId/evidencexpert.DoIntegratedSearch?Search Term=nortriptyline (accessed 30 April 2014).

33. Anderson IM, Ferrier IN, Baldwin RC, et al. Evidence- based guidelines for treating depressive disorders with anti- depressants: a revision of the 2000 British Association for

 

 

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

26

 

Prosthetics and Orthotics International 39(1)

 

 

page10image1352

Psychopharmacology guidelines. J Psychopharmacol 2008;

22(4): 343–396.

  1. Waring WS. Clinical use of antidepressant therapy and

    associated cardiovascular risk. Drug Healthc Patient Saf

    2012; 4: 93–101.

  2. Cohen HW, Gibson G and Alderman MH. Excess risk of

    myocardial infarction in patients treated with antidepressant medications: association with use of tricyclic agents. Am J Med 2000; 108: 2–8.

  3. US Food and Drug Administration. Medication guide: antide- pressant medicines, depression and other serious mental ill- nesses, and suicidal thoughts or actions, http://www.fda.gov/ downloads/Drugs/DrugSafety/InformationbyDrugClass/ ucm100211.pdf

  4. US Food and Drug Administration. Medication guide: Pamelor, http://www.fda.gov/downloads/Drugs/Drug Safety/ucm088671.pdf (accessed 27 April 2014).

  5. Morello CM, Leckband SG, Stoner CP, et al. Randomized double-blind study comparing the efficacy of gabapentin with amitriptyline on diabetic peripheral neuropathy pain. Arch Intern Med 1999; 159: 1931–1937.

  6. Marks DM, Shah MJ, Patkar AA, et al. Serotonin- norepinephrine reuptake inhibitors for pain control: premise and promise. Curr Neuropharmacol 2009; 7: 331–336.

  7. Saarto T and Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev 2007; 4: CD005454.

  8. Lunn MP, Hughes RA and Wiffen PJ. Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia. Cochrane Database Syst Rev 2014; 1: CD007115.

  9. Brecht S, Courtecuisse C, Debieuvre C, et al. Efficacy and safety of duloxetine 60mg once daily in the treatment of pain in patients with major depressive disorder and at least moderate pain of unknown etiology: a randomized con- trolled trial. J Clin Psychiatry 2007; 68: 1707–1716.

  10. Serebruany VL. Selective serotonin reuptake inhibitors and increased bleeding risk: are we missing something? Am J Med 2006; 119(2): 113–136.

  11. US Food and Drug Administration. Medication guide: Cymbalta http://www.fda.gov/downloads/Drugs/Drug Safety/ucm088579.pdf (accessed 28 April 2014).

  12. Kaur H, Hota D, Bhansali A, et al. A comparative evalu- ation of amitriptyline and duloxetine in painful diabetic neuropathy: a randomized, double-blind, cross-over clinical trial. Diabetes Care 2011; 34: 818–822.

  13. Rose MA and Kam PC. Gabapentin: pharmacology and its use in pain management. Anaesthesia 2002 May; 57: 451–462.

  14. Fehrenbacher JC, Taylor CP and Vasko MR. Pregabalin and gabapentin reduce release of substance P and CGRP from rat spinal tissues only after inflammation or activation of protein kinase C. Pain 2003; 105: 133–141.

  15. US Food and Drug Administration. Medication guide: Lyrica, http://www.fda.gov/downloads/Drugs/DrugSafety/ UCM152825.pdf (accessed 28 April 2014).

  16. US Food and Drug Administration. Medication guide: Neurontin, http://www.fda.gov/downloads/Drugs/ DrugSafety/UCM229208.pdf (accessed 28 April 2014).

  17. Jensen MP, Chodroff MJ and Dworkin RH. The impact of neuropathic pain on health-related quality of life: review and implications. Neurology 2007; 68: 1178–1182.

51. Pollack MH, Matthews J and Scott EL. Gabapentin as a potential treatment for anxiety disorders. Am J Psychiatry 1998; 155: 992–993.

52. Lesser H, Sharma U, LaMoreaux L, et al. Pregabalin relieves symptoms of painful diabetic neuropathy: a rand- omized controlled trial. Neurology 2004; 63: 2104–2110.

53. Wong MC, Chung JW and Wong TK. Effects of treatments for symptoms of painful diabetic neuropathy: systematic review. BMJ 2007; 335: 87.

54. Afilalo M and Morlion B. Efficacy of tapentadol ER for managing moderate to severe chronic pain. Pain Physician 2013; 16: 27–40.

55. Sindrup SH, Andersen G, Madsen C, et al. Tramadol relieves pain and allodynia in polyneuropathy: a randomised, dou- ble-blind, controlled trial. Pain 1999; 83: 85–90.

56. Harati Y, Gooch C, Swenson M, et al. Double-blind ran- domized trial of tramadol for the treatment of the pain of diabetic neuropathy. Neurology 1998; 50: 1842–1846.

57. Janssen Pharmaceuticals. NDA 200533: NUCYNTA ER Tablets (Opioid analgesics), http://www.fda.gov/down- loads/drugs/drugsafety/postmarketdrugsafetyinforma- tionforpatientsandproviders/ucm270224.pdf (accessed 25 August 2011).

58. Raffa RB, Buschmann H, Christoph T, et al. Mechanistic and functional differentiation of tapentadol and tramadol. Expert Opin Pharmacother 2012; 13: 1437–1449.

59. Niesters M, Proto PL, Aarts L, et al. Tapentadol potentiates descending pain inhibition in chronic pain patients with dia- betic polyneuropathy. Br J Anaesth 2014; 113: 148–156.

60. Kosten TR and George TP. The neurobiology of opioid dependence: implications for treatment. Sci Pract Perspect 2002; 1: 13–20.

61. National Survey on Drug Use and Health (NSDUH) is an annual survey sponsored by the Substance Abuse and Mental Health Services Administration (SAMHSA), http:// oas.samhsa.gov

62. Savage SR. Assessment for addiction in pain-treatment set- tings. Clin J Pain 2002; 18: S28–S38.

63. Benyamin R, Trescot AM, Datta S, et al. Opioid complica- tions and side effects. Pain Physician 2008; 11: S105–S120. 64. Dean M. Opioids in renal failure and dialysis patients. J

Pain Symptom Manage 2004; 28: 497–504.
65. Dwyer JP, Jayasekera C and Nicoll A. Analgesia for the

cirrhotic patient: a literature review and recommendations. J Gastroenterol Hepatol. Epub ahead of print 18 February 2014. DOI: 10.1111/jgh.12560.

66. US Food and Drug Administration. NUCYNTA (tap- entadol) immediate-release oral tablets: safety labeling changes approved by FDA Center for Drug Evaluation and Research (CDER), October 2013, http://www.fda.gov/ Safety/MedWatch/SafetyInformation/ucm233891.htm

67. US Food and Drug Administration. NUCYNTA ER FDA risk evaluation and mitigation strategy, approved 25 August 2011, http://www.fda.gov/downloads/drugs/drugsafety/ postmarketdrugsafetyinformationforpatientsandproviders/ ucm270224.pdf

68. US Food and Drug Administration. Ultram ER pre- scribing information, updated September 2006, http://www.accessdata.fda.gov/drugsatfda_docs/ label/2008/021692s005s007lbl.pdf

 

 

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

Rosenberg 27

 

 

page11image880

  1. Gimbel JS, Richards P and Portenoy RK. Controlled-release oxycodone for pain in diabetic neuropathy: a randomized controlled trial. Neurology 2003; 60: 927–934.

  2. Watson CP, Moulin D, Watt-Watson J, et al. Controlled- release oxycodone relieves neuropathic pain: a randomized controlled trial in painful diabetic neuropathy. Pain 2003; 105: 71–78.

  3. Rowbotham MC, Twilling L, Davies PS, et al. Oral opioid therapy for chronic peripheral and central neuropathic pain. N Engl J Med 2003; 348: 1223–1232.

  4. Schwartz S, Etropolski M, Shapiro DY, et al. Safety and efficacy of tapentadol ER in patients with painful diabetic peripheral neuropathy: results of a randomized-withdrawal, placebo-controlled trial. Curr Med Res Opin 2011; 27: 151–162.

  5. Raja SN, Haythornthwaite JA, Pappagallo M, et al. Opioids versus antidepressants in postherpetic neuralgia: a ran- domized, placebo-controlled trial. Neurology 2002; 59: 1015–1021.

  6. Nuckols TK, Anderson L, Popescu I, et al. Opioid prescrib- ing: a systematic review and critical appraisal of guidelines for chronic pain. Ann Intern Med 2014; 160: 38–47.

  7. Mayo Clinic. Capsaicin (topical route): side effects, http:// www.mayoclinic.org/drugs-supplements/capsaicin-topical- route/side-effects/drg-20062561 (accessed 27 April 2014).

  8. Qutenza. Healthcare professional resources: safety informa- tion, http://www.qutenza.com/ (accessed 27 April 2014).

  9. Simpson DM, Gazda S, Brown S, et al. Long-term safety of NGX-4010, a high-concentration capsaicin patch, in patients with peripheral neuropathic pain. J Pain Symptom Manage 2010; 39: 1053–1064.

  10. Derry S, Sven-Rice A, Cole P, et al. Topical capsaicin (high concentration) for chronic neuropathic pain in adults. Cochrane Database Syst Rev 2013; 2: CD007393.

  11. Cummins TR. Setting up for the block: the mechanism underlying lidocaine’s use-dependent inhibition of sodium channels. J Physiol 2007; 582(Pt 1): 11.

  12. Endo Pharmaceuticals, Inc. LIDODERM lidocaine patch 5%, http://www.endo.com/File%20Library/Products/ Prescribing%20Information/LIDODERM_prescribing_ information.html (accessed 30 April 2014).

  13. Barbano RL, Herrmann DN, Hart-Gouleau S, et al. Effectiveness, tolerability, and impact on quality of life of the 5% lidocaine patch in diabetic polyneuropathy. Arch Neurol 2004; 61: 914–918.

  14. Baron R, Mayoral V, Leijon G, et al. Efficacy and safety of combination therapy with 5% lidocaine medicated plaster and pregabalin in post-herpetic neuralgia and diabetic pol- yneuropathy. Curr Med Res Opin 2009; 258: 1677–1687.

  15. Proudfoot CJ, Garry EM, Cottrell DF, et al. Analgesia medi- ated by the TRPM8 cold receptor in chronic neuropathic pain. Curr Biol 2006; 16: 1591–1605.

  16. Davies SJ, Harding LM and Baranowski AP. A novel treat- ment of postherpetic neuralgia using peppermint oil. Clin J Pain 2002; 18: 200–202.

  17. Wasner G, Naleschinski D, Binder A, et al. The effect of menthol on cold allodynia in patients with neuropathic pain. Pain Med 2008 9; 354–358.

  18. Lynch ME, Clark AJ, Sawynok J, et al. Topical 2% ami- triptyline and 1% ketamine in neuropathic pain syndromes:

a randomized, double-blind, placebo-controlled trial.

Anesthesiology 2005; 103: 140–146.
87. Lynch ME, Clark AJ, Sawynok J, et al. Topical amitripty-

line and ketamine in neuropathic pain syndromes: an open-

label study. J Pain 2005; 6: 644–649.
88. Gewandter JS, Mohile SG, Heckler CE, et al. A phase III

randomized, placebo-controlled study of topical amitripty- line and ketamine for chemotherapy-induced peripheral neu- ropathy (CIPN): a University of Rochester CCOP study of 462 cancer survivors. Support Care Cancer. Epub ahead of print 16 February 2014. DOI: 10.1007/s00520-014-2158-7.

89. Barton DL, Wos EJ, Qin R, et al. A double-blind, placebo- controlled trial of a topical treatment for chemotherapy- induced peripheral neuropathy: NCCTG trial N06CA. Support Care Cancer 2011; 19: 833–841.

90. Nagamatsu M, Nickander KK, Schmelzer JD, et al. Lipoic acid improves nerve blood flow, reduces oxidative stress, and improves distal nerve conduction in experimental dia- betic neuropathy. Diabetes Care 1995; 18: 1160–1167.

91. Ziegler D, Ametov A, Barinov A, et al. Oral treatment with alpha-lipoic acid improves symptomatic diabetic polyneuropa- thy: the SYDNEY 2 trial. Diabetes Care 2006; 29: 2365–2370.

92. Ziegler D, Nowak H, Kempler P, et al. Treatment of symp- tomatic diabetic polyneuropathy with the antioxidant alpha- lipoic acid: a meta-analysis. Diabet Med 2004; 21: 114–121.

93. Scafidi S, Fiskum G, Lindauer SL, et al. Metabolism of acetyl-l-carnitine for energy and neurotransmitter synthesis in the immature rat brain. J Neurochem 2010; 114: 820–831.

94. Evans JD, Jacobs TF and Evans EW. Role of acetyl-l-carni- tine in the treatment of diabetic peripheral neuropathy. Ann Pharmacother 2008; 42: 1686–1691.

95. Sima AA. Acetyl-l-carnitine in diabetic polyneuropathy: experimental and clinical data. CNS Drugs 2007; 21: 13–23. 96. Sima AA, Calvani M, Mehra M, et al. Acetyl-l-carnitine improves pain, nerve regeneration, and vibratory perception in patients with chronic diabetic neuropathy: an analysis of two randomized

placebo-controlled trials. Diabetes Care 2005; 28: 89–94.
97. Abuaisha BB, Costanzi JB and Boulton AJ. Acupuncture for the treatment of chronic painful peripheral diabetic neu- ropathy: a long-term study.
Diabetes Res Clin Pract 1998;

39: 115–121.
98. Jiang H, Shi K, Li X, et al. Clinical study on the wrist-ankle

acupuncture treatment for 30 cases of diabetic peripheral

neuritis. J Tradit Chin Med 2006; 26: 8–12.
99. Tong Y, Guo H and Han B. Fifteen-day acupuncture treat- ment relieves diabetic peripheral neuropathy.
J Acupunct

Meridian Stud 2010; 3: 95–103.
100. Dubinsky RM and Miyasaki J. Assessment: efficacy of

transcutaneous electric nerve stimulation in the treatment of pain in neurologic disorders (an evidence-based review): report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology 2010; 74: 173–176.

101. Kumar D and Marshall HJ. Diabetic peripheral neuropathy: amelioration of pain with transcutaneous electrostimulation. Diabetes Care 1997; 20: 1702–1705.

102. Forst T, Nguyen M, Forst S, et al. Impact of low frequency transcutaneous electrical nerve stimulation on sympto- matic diabetic neuropathy using the new Salutaris device. Diabetes Nutr Metab 2004; 17: 163–168.

 

 

Downloaded from poi.sagepub.com by guest on July 3, 2015

 

 

28

 

Prosthetics and Orthotics International 39(1)

 

 

page12image1352

  1. Glombiewski JA, Hartwich-Tersek J and Rief W. Two psy- chological interventions are effective in severely disabled, chronic back pain patients: a randomised controlled trial. Int J Behav Med 2010; 17: 97–107.

  2. Hoffman BM, Papas RK, Chatkoff DK, et al. Meta-analysis of psychological interventions for chronic low back pain. Health Psychol 2007; 26: 1–9.

  3. Holroyd KA, O’Donnell FJ, Stensland M, et al. Management of chronic tension-type headache with tricyclic antidepres- sant medication, stress management therapy, and their combination: a randomized controlled trial. JAMA 2001; 285: 2208–2215.

  4. Otis JD, Sanderson K, Hardway C, et al. A randomized controlled pilot study of a cognitive-behavioral therapy approach for painful diabetic peripheral neuropathy. J Pain 2013; 14: 475–482.

  5. Daousi C, Benbow SJ and MacFarlane IA. Electrical spinal cord stimulation in the long-term treatment of chronic painful diabetic neuropathy. Diabet Med 2005; 22: 393–398.

108. Tesfaye S, Watt J, Benbow SJ, et al. Electrical spinal-cord stimulation for painful diabetic peripheral neuropathy. Lancet 1996; 348: 1698–1701.

109. Kumar K, Toth C and Nath RK. Spinal cord stimulation for chronic pain in peripheral neuropathy. Surg Neurol 1996; 46: 363–369.

110. Huntoon MA and Burgher AH. Ultrasound-guided perma- nent implantation of peripheral nerve stimulation (PNS) system for neuropathic pain of the extremities: original cases and outcomes. Pain Med 2009; 10: 1369–1377.

111. Medtronic Targeted Drug Delivery. Indications, safety, and warnings, http://professional.medtronic.com/pt/neuro/idd/ ind/ (accessed 27 April 2014).

112. Miljanich GP. Ziconotide: neuronal calcium channel blocker for treating severe chronic pain. Curr Med Chem 2004; 11: 3029–3040.

113. Deer TR, Smith HS, Cousins M, et al. Consensus guidelines for the selection and implantation of patients with noncan- cer pain for intrathecal drug delivery. Pain Physician 2010; 13: E175–E213.

 

 

Downloaded from poi.sagepub.com by guest on July 3, 2015 

 

 

Comments

comments

Author

PV Mayer

Dr. Perry Mayer is the Medical Director of The Mayer Institute (TMI), a center of excellence in the treatment of the diabetic foot. He received his undergraduate degree from Queen’s University, Kingston and medical degree from the Royal College of Surgeons in Ireland.

Add Comment