542266 research-article2014

POI0010.1177/0309364614542266Prosthetics and Orthotics InternationalRosenberg

INTERNATIONAL SOCIETY FOR PROSTHETICS AND ORTHOTICS

Literature Review

Treatment of painful diabetic peripheral neuropathy

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

Casandra J Rosenberg1 and James C Watson2

Abstract 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 population.1 Up to two-thirds of people with diabetes develop peripheral neuropathy, with an increasing prevalence with longer disease duration; 15%–25% of diabetic neuropathies 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 multiple 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 person with painful DPN is challenging.12 Choosing a treatment 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 initiated, 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 peripheral 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]

Downloaded from poi.sagepub.com at SAN DIEGO STATE UNIV LIBRARY on January 27, 2015

18

Prosthetics and Orthotics International 39(1)

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-effectiveness, while the NeuPSIG and the American Academy of Neurology (AAN) also consider quality-of-life outcomes. One of the guidelines is strictly for painful DPN,20 and three others include specific subgroup recommendations for DPN.16,18,19

Disease management The first step in the management of painful DPN is treating the cause of neuropathy, the diabetes. There is evidence 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 developing DPN of 0.35 for patients who undergo intensive glucose control compared to conventional treatment (95% confidence interval (CI) = 0.23–0.53; p 400 mg/day

Sedation, confusion, dizziness, peripheral edema, euphoria

Sedation, confusion, dizziness, peripheral edema, tremor

Fatigue, nausea, hyperhidrosis

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

Most common side effects

Nausea, rash, and hypothyroidism

No maximum Constipation, nausea/ dosage vomiting, drowsiness, confusion, dizziness, pruritus

600 mg/day

400 mg/day

600 mg/day

3600 mg/day (divided into three doses)

60 mg twice daily

Maximum dosage

Dosage and titration recommendations

Agent

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

Hypoglycemia

Renal insufficiency (must dose adjust), psychiatric disease (euphoria risk), withdrawal syndromes/seizure with abrupt discontinuation History of substance abuse, sedation, driving impairment, seizure disorder, risk of serotonin syndrome with concomitant use of SSRI, SNRI, and TCAs 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 History of substance abuse, driving impairment, tolerance, abuse, diversion

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 Small risk of emerging suicidality (boxed warning), increased bleeding risk (care with anticoagulants), withdrawal syndromes with abrupt discontinuation, serotonin syndrome, hepatic failure Renal insufficiency (must dose adjust), withdrawal syndromes/seizure with abrupt discontinuation

Precautions

Few toxicities

Rapid onset analgesic benefit

Rapid onset analgesic benefit

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

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

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

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

Other benefits

22 Prosthetics and Orthotics International 39(1)

23

Rosenberg

No known systemic toxicities

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

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

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

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

May apply up to four patches at one time. 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. Apply cream to affected areas four times daily Topical capsaicin (high concentration 8% patch)

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

Low risk of systemic toxicity

No known systemic toxicities Unknown risk from repeated exposures, not recommended for face or over broken skin

Few toxicities 1000 mg three times per day Acetyl-l-carnitine

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

Maximum dosage Dosage and titration recommendations Agent

Table 1. (Continued)

Most common side effects

Precautions

Other benefits

Other supplements

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) delivers an electrical current via electrodes on the skin arranged about the affected area. The intensity, frequency, and pattern of stimulation can be adjusted by the patient or therapist. Several studies including sham treatment arms have demonstrated benefit in painful DPN.100–102

Cognitive behavioral therapy Cognitive behavioral therapy (CBT) replaces dysfunctional thoughts and behaviors with positive ones and has been found effective in multiple pain states.103–105 One randomized pilot study showed benefit for patients with painful 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 targeted areas, or ablation/modulation of targeted nerves.”13 Recently, the NeuPSIG compiled evidence for interventional treatments in the management of neuropathic pain, including a subgroup analysis for painful DPN in combination with other painful peripheral neuropathies.

Neuromodulation Neuromodulation involves the application of electrical current to neural structures to modulate neural transmission.

Downloaded from poi.sagepub.com at SAN DIEGO STATE UNIV LIBRARY on January 27, 2015

24

Prosthetics and Orthotics International 39(1)

The most common method of neuromodulation for pain is dorsal column stimulation, also known as spinal cord stimulation (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 implantation, 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 traditional 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 tunnelled into the cerebrospinal fluid (CSF). Morphine and ziconotide (a selective N-type voltage-gated calcium channel blocker) are FDA-approved medications for intrathecal infusion.111,112 There are currently no studies showing evidence for intrathecal drug delivery specifically for neuropathic noncancer 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 intrathecal system need to be able to maintain regular follow-up for system refills and adjustments.

Managing expectations is vital when starting or changing medications. Most treatments are considered successful 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 serious side effects should be had prior to starting a new treatment. Many of these treatment side effects can be improved over time due to progressive tolerance; these can be mitigated 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

Conclusion While painful DPN can prove difficult to treat, there are a variety of therapeutic options, including behavioral therapies, 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 medical therapy. Many painful DPN treatments can also address other comorbid conditions. Furthermore, some medication side effects could exacerbate preexisting problems and should be avoided.

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 diabetes 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 at SAN DIEGO STATE UNIV LIBRARY on January 27, 2015

25

Rosenberg 3. 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. 4. 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. 5. 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. 6. 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. 7. Berger A, Dukes EM and Oster G. Clinical characteristics and economic costs of patients with painful neuropathic disorders. J Pain 2004; 5: 143–149. 8. 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. 9. Benbow SJ, Wallymahmed ME and MacFarlane IA. Diabetic peripheral neuropathy and quality of life. QJM 1998; 91: 733–737. 10. Galer BS, Gianas A and Jensen MP. Painful diabetic polyneuropathy: epidemiology, pain description, and quality of life. Diabetes Res Clin Pract 2000; 47: 123–128. 11. 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. 12. Kingery WS. A critical review of controlled clinical trials for peripheral neuropathic pain and complex regional pain syndromes. Pain 1997; 73: 123–139. 13. Dworkin RH. Inclusion criteria, http://www.acc.co.nz/ for-providers/clinical-best-practice/interventional-painmanagement/interventions/intervention-index/ WCM1_034233 (2013, accessed 27 March 2014). 14. O’Connor AB and Dworkin RH. Treatment of neuropathic pain: an overview of recent guidelines. Am J Med 2009; 122: S22–S32. 15. Davies HT, Crombie IK, Lonsdale M, et al. Consensus and contention in the treatment of chronic nerve-damage pain. Pain 1991; 47: 191–196. 16. Finnerup NB, Otto M, McQuay HJ, et al. Algorithm for neuropathic pain treatment: an evidence based proposal. Pain 2005; 118: 289–305. 17. Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol 2010; 17: 1113–e88. 18. 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. 19. Moulin DE, Clark AJ, Gilron I, et al. Pharmacological management of chronic neuropathic pain—consensus statement

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 control 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 importance of changes in chronic pain intensity measured on an 11-point numerical pain rating scale. Pain 2001; 94: 149– 158. 26. Gilron I, Bailey JM, Tu D, et al. Nortriptyline and gabapentin, 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 antidepressants 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/evidencexpert/CS/54AC7A/ND_AppProduct/evidencexpert/ DUPLICATIONSHIELDSYNC/AEFE69/ND_PG/evidencexpert/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/evidencexpert/CS/B42175/ND_AppProduct/evidencexpert/ DUPLICATIONSHIELDSYNC/56E6AD/ND_PG/evidencexpert/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. Evidencebased guidelines for treating depressive disorders with antidepressants: a revision of the 2000 British Association for

Downloaded from poi.sagepub.com at SAN DIEGO STATE UNIV LIBRARY on January 27, 2015

26

Prosthetics and Orthotics International 39(1)

Psychopharmacology guidelines. J Psychopharmacol 2008; 22(4): 343–396. 34. Waring WS. Clinical use of antidepressant therapy and associated cardiovascular risk. Drug Healthc Patient Saf 2012; 4: 93–101. 35. 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. 36. US Food and Drug Administration. Medication guide: antidepressant medicines, depression and other serious mental illnesses, and suicidal thoughts or actions, http://www.fda.gov/ downloads/Drugs/DrugSafety/InformationbyDrugClass/ ucm100211.pdf 37. US Food and Drug Administration. Medication guide: Pamelor, http://www.fda.gov/downloads/Drugs/Drug Safety/ucm088671.pdf (accessed 27 April 2014). 38. 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. 39. Marks DM, Shah MJ, Patkar AA, et al. Serotonin norepinephrine reuptake inhibitors for pain control: premise and promise. Curr Neuropharmacol 2009; 7: 331–336. 40. Saarto T and Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev 2007; 4: CD005454. 41. Lunn MP, Hughes RA and Wiffen PJ. Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia. Cochrane Database Syst Rev 2014; 1: CD007115. 42. Brecht S, Courtecuisse C, Debieuvre C, et al. Efficacy and safety of duloxetine 60 mg once daily in the treatment of pain in patients with major depressive disorder and at least moderate pain of unknown etiology: a randomized controlled trial. J Clin Psychiatry 2007; 68: 1707–1716. 43. Serebruany VL. Selective serotonin reuptake inhibitors and increased bleeding risk: are we missing something? Am J Med 2006; 119(2): 113–136. 44. US Food and Drug Administration. Medication guide: Cymbalta http://www.fda.gov/downloads/Drugs/Drug Safety/ucm088579.pdf (accessed 28 April 2014). 45. Kaur H, Hota D, Bhansali A, et al. A comparative evaluation of amitriptyline and duloxetine in painful diabetic neuropathy: a randomized, double-blind, cross-over clinical trial. Diabetes Care 2011; 34: 818–822. 46. Rose MA and Kam PC. Gabapentin: pharmacology and its use in pain management. Anaesthesia 2002 May; 57: 451–462. 47. 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. 48. US Food and Drug Administration. Medication guide: Lyrica, http://www.fda.gov/downloads/Drugs/DrugSafety/ UCM152825.pdf (accessed 28 April 2014). 49. US Food and Drug Administration. Medication guide: Neurontin, http://www.fda.gov/downloads/Drugs/ DrugSafety/UCM229208.pdf (accessed 28 April 2014). 50. 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 randomized 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, double-blind, controlled trial. Pain 1999; 83: 85–90. 56. Harati Y, Gooch C, Swenson M, et al. Double-blind randomized 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/downloads/drugs/drugsafety/postmarketdrugsafetyinformationforpatientsandproviders/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 diabetic 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 settings. Clin J Pain 2002; 18: S28–S38. 63. Benyamin R, Trescot AM, Datta S, et al. Opioid complications 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 at SAN DIEGO STATE UNIV LIBRARY on January 27, 2015

27

Rosenberg 69. Gimbel JS, Richards P and Portenoy RK. Controlled-release oxycodone for pain in diabetic neuropathy: a randomized controlled trial. Neurology 2003; 60: 927–934. 70. Watson CP, Moulin D, Watt-Watson J, et al. Controlledrelease oxycodone relieves neuropathic pain: a randomized controlled trial in painful diabetic neuropathy. Pain 2003; 105: 71–78. 71. 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. 72. 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. 73. Raja SN, Haythornthwaite JA, Pappagallo M, et al. Opioids versus antidepressants in postherpetic neuralgia: a randomized, placebo-controlled trial. Neurology 2002; 59: 1015–1021. 74. Nuckols TK, Anderson L, Popescu I, et al. Opioid prescribing: a systematic review and critical appraisal of guidelines for chronic pain. Ann Intern Med 2014; 160: 38–47. 75. Mayo Clinic. Capsaicin (topical route): side effects, http:// www.mayoclinic.org/drugs-supplements/capsaicin-topicalroute/side-effects/drg-20062561 (accessed 27 April 2014). 76. Qutenza. Healthcare professional resources: safety information, http://www.qutenza.com/ (accessed 27 April 2014). 77. 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. 78. 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. 79. 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. 80. Endo Pharmaceuticals, Inc. LIDODERM lidocaine patch 5%, http://www.endo.com/File%20Library/Products/ Prescribing%20Information/LIDODERM_prescribing_ information.html (accessed 30 April 2014). 81. 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. 82. 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 polyneuropathy. Curr Med Res Opin 2009; 258: 1677–1687. 83. Proudfoot CJ, Garry EM, Cottrell DF, et al. Analgesia mediated by the TRPM8 cold receptor in chronic neuropathic pain. Curr Biol 2006; 16: 1591–1605. 84. Davies SJ, Harding LM and Baranowski AP. A novel treatment of postherpetic neuralgia using peppermint oil. Clin J Pain 2002; 18: 200–202. 85. 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. 86. Lynch ME, Clark AJ, Sawynok J, et al. Topical 2% amitriptyline 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 amitriptyline and ketamine in neuropathic pain syndromes: an openlabel 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 amitriptyline and ketamine for chemotherapy-induced peripheral neuropathy (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, placebocontrolled trial of a topical treatment for chemotherapyinduced 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 diabetic 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 polyneuropathy: the SYDNEY 2 trial. Diabetes Care 2006; 29: 2365–2370. 92. Ziegler D, Nowak H, Kempler P, et al. Treatment of symptomatic diabetic polyneuropathy with the antioxidant alphalipoic 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-carnitine 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 neuropathy: 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 treatment 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 symptomatic diabetic neuropathy using the new Salutaris device. Diabetes Nutr Metab 2004; 17: 163–168.

Downloaded from poi.sagepub.com at SAN DIEGO STATE UNIV LIBRARY on January 27, 2015

28

Prosthetics and Orthotics International 39(1)

103. Glombiewski JA, Hartwich-Tersek J and Rief W. Two psychological interventions are effective in severely disabled, chronic back pain patients: a randomised controlled trial. Int J Behav Med 2010; 17: 97–107. 104. Hoffman BM, Papas RK, Chatkoff DK, et al. Meta-analysis of psychological interventions for chronic low back pain. Health Psychol 2007; 26: 1–9. 105. Holroyd KA, O’Donnell FJ, Stensland M, et al. Management of chronic tension-type headache with tricyclic antidepressant medication, stress management therapy, and their combination: a randomized controlled trial. JAMA 2001; 285: 2208–2215. 106. 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. 107. 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 permanent 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 noncancer pain for intrathecal drug delivery. Pain Physician 2010; 13: E175–E213.

Downloaded from poi.sagepub.com at SAN DIEGO STATE UNIV LIBRARY on January 27, 2015

Treatment of painful diabetic peripheral neuropathy.

Painful diabetic peripheral neuropathy impairs quality of life and can be difficult to treat...
540KB Sizes 4 Downloads 17 Views