190
Views
0
CrossRef citations to date
0
Altmetric
Neuropathic Pain

A Systematic Guideline by the ASPN Workgroup on the Evidence, Education, and Treatment Algorithm for Painful Diabetic Neuropathy: SWEET

ORCID Icon, ORCID Icon, ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, , ORCID Icon, ORCID Icon, , ORCID Icon, , ORCID Icon, , ORCID Icon, ORCID Icon, , , ORCID Icon, , ORCID Icon, , ORCID Icon & show all
Pages 1461-1501 | Received 21 Nov 2023, Accepted 19 Mar 2024, Published online: 12 Apr 2024

References

  • Sayed D, Grider J, Strand N, et al. The American Society of Pain and Neuroscience (ASPN) Evidence-Based Clinical Guideline of Interventional Treatments for Low Back Pain. J Pain Res. 2022;15:3729–3832. doi:10.2147/JPR.S386879
  • Harris RP, Helfand M, Woolf SH, et al. Current methods of the U.S. preventive services task force: a review of the process. Am J Preventive Med. 2001;20(3 Suppl):21–35. doi:10.1016/S0749-3797(01)00261-6
  • Daousi C, MacFarlane IA, Woodward A, Nurmikko TJ, Bundred PE, Benbow SJ. Chronic painful peripheral neuropathy in an urban community: a controlled comparison of people with and without diabetes. Diabet Med. 2004;21(9):976–982. doi:10.1111/j.1464-5491.2004.01271.x
  • 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(3):206–213. doi:10.1016/j.diabet.2008.11.004
  • Tesfaye S, Boulton AJM, Dyck PJ, et al. Diabetic neuropathies: update on definitions, diagnostic criteria, estimation of severity, and treatments. Diabetes Care. 2010;33(10):2285–2293. doi:10.2337/dc10-1303
  • Dowell D, Haegerich TM, Chou R. CDC Guideline for Prescribing Opioids for Chronic Pain--United States, 2016. JAMA. 2016;315(15):1624–1645. doi:10.1001/jama.2016.1464
  • Franklin GM; American Academy of Neurology. Opioids for chronic noncancer pain: a position paper of the American Academy of Neurology. Neurology. 2014;83(14):1277–1284. doi:10.1212/WNL.0000000000000839
  • Callaghan BC, Reynolds E, Banerjee M, Kerber KA, Skolarus LE, Burke JF. Longitudinal pattern of pain medication utilization in peripheral neuropathy patients. Pain. 2019;160(3):592–599. doi:10.1097/j.pain.0000000000001439
  • Castelli G, Desai KM, Cantone RE. Peripheral Neuropathy: evaluation and Differential Diagnosis. Am Fam Physician. 2020;102(12):732–739.
  • Pop-Busui R, Boulton AJM, Feldman EL, et al. Diabetic Neuropathy: a Position Statement by the American Diabetes Association. Diabetes Care. 2017;40(1):136–154. doi:10.2337/dc16-2042
  • Meijer JWG, Smit AJ, Lefrandt JD, van der Hoeven JH, Hoogenberg K, Links TP. Back to basics in diagnosing diabetic polyneuropathy with the tuning fork! Diabetes Care. 2005;28(9):2201–2205. doi:10.2337/diacare.28.9.2201
  • Feng Y, Schlösser FJ, Sumpio BE. The Semmes Weinstein monofilament examination as a screening tool for diabetic peripheral neuropathy. J Vascular Surg. 2009;50(3):675–682.e1. doi:10.1016/j.jvs.2009.05.017
  • Vlckova-Moravcova E, Bednarik J, Belobradkova J, Sommer C. Small-fibre involvement in diabetic patients with neuropathic foot pain. Diabet Med. 2008;25(6):692–699. doi:10.1111/j.1464-5491.2008.02446.x
  • Chan AW, MacFarlane IA, Bowsher D, Campbell JA. Weighted needle pinprick sensory thresholds: a simple test of sensory function in diabetic peripheral neuropathy. J Neurol Neurosurg Psychiatry. 1992;55(1):56–59. doi:10.1136/jnnp.55.1.56
  • Li ZF, Niu XL, Nie LL, Chen LP, Cao CF, Guo L. Diagnostic value of clinical deep tendon reflexes in diabetic peripheral neuropathy. Arch Med Sci. 2023;19(5):656. doi:10.5114/aoms.2020.100656
  • Maras O, Dulgeroglu D, Cakci A. Ankle Proprioception in Patients with Type 2 Diabetes Mellitus. J Am Podiatr Med Assoc. 2021;111(4):8. doi:10.7547/18-178
  • England JD, Gronseth GS, Franklin G, et al. Practice Parameter: evaluation of distal symmetric polyneuropathy: role of laboratory and genetic testing (an evidence-based review). Report of the American Academy of Neurology, American Association of Neuromuscular and Electrodiagnostic Medicine, and American Academy of Physical Medicine and Rehabilitation. Neurology. 2009;72(2):185–192. doi:10.1212/01.wnl.0000336370.51010.a1
  • Timar B, Popescu S, Timar R, et al. The usefulness of quantifying intraepidermal nerve fibers density in the diagnostic of diabetic peripheral neuropathy: a cross-sectional study. Diabetol Metab Syndr. 2016;8:31. doi:10.1186/s13098-016-0146-4
  • Beiswenger KK, Calcutt NA, Mizisin AP. Epidermal nerve fiber quantification in the assessment of diabetic neuropathy. Acta Histochem. 2008;110(5):351–362. doi:10.1016/j.acthis.2007.12.004
  • Price R, Smith D, Franklin G, et al. Oral and Topical Treatment of Painful Diabetic Polyneuropathy: practice Guideline Update Summary: report of the AAN Guideline Subcommittee. Neurology. 2022;98(1):31–43. doi:10.1212/WNL.0000000000013038
  • Cymbalta Package Insert; 2017. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021427s049lbl.pdf. Accessed June 24, 2023.
  • Lyrica Package Insert. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/021446s035,022488s013lbl.pdf. Accessed June 24, 2023.
  • Nucynta Package Insert; 2016. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/200533s014lbl.pdf. Accessed June 24, 2023.
  • D’Souza RS, Barman R, Joseph A, Abd-Elsayed A. Evidence-Based Treatment of Painful Diabetic Neuropathy: a Systematic Review. Curr Pain Headache Rep. 2022;26(8):583–594. doi:10.1007/s11916-022-01061-7
  • 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(1):151–162. doi:10.1185/03007995.2010.537589
  • Tesfaye S, Wilhelm S, Lledo A, et al. Duloxetine and pregabalin: high-dose monotherapy or their combination? The “COMBO-DN study”--a multinational, randomized, double-blind, parallel-group study in patients with diabetic peripheral neuropathic pain. Pain. 2013;154(12):2616–2625. doi:10.1016/j.pain.2013.05.043
  • Marks DM, Shah MJ, Patkar AA, Masand PS, Park GY, Pae CU. Serotonin-norepinephrine reuptake inhibitors for pain control: premise and promise. Curr Neuropharmacol. 2009;7(4):331–336. doi:10.2174/157015909790031201
  • Raskin J, Wang F, Pritchett YL, Goldstein DJ. Duloxetine for patients with diabetic peripheral neuropathic pain: a 6-month open-label safety study. Pain Med. 2006;7(5):373–385. doi:10.1111/j.1526-4637.2006.00207.x
  • Goldstein DJ, Lu Y, Detke MJ, Lee TC, Iyengar S. Duloxetine vs. placebo in patients with painful diabetic neuropathy. Pain. 2005;116(1–2):109–118. doi:10.1016/j.pain.2005.03.029
  • Neurontin Package Insert; 2017. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/020235s041,020882s028,021129s027lbl.pdf. Accessed June 24, 2023.
  • Bockbrader HN, Wesche D, Miller R, Chapel S, Janiczek N, Burger P. A comparison of the pharmacokinetics and pharmacodynamics of pregabalin and gabapentin. Clin Pharmacokinet. 2010;49(10):661–669. doi:10.2165/11536200-000000000-00000
  • Backonja M, Beydoun A, Edwards KR, et al. Gabapentin for the symptomatic treatment of painful neuropathy in patients with diabetes mellitus: a randomized controlled trial. JAMA. 1998;280(21):1831–1836. doi:10.1001/jama.280.21.1831
  • Dallocchio C, Buffa C, Mazzarello P, Chiroli S. Gabapentin vs. amitriptyline in painful diabetic neuropathy: an open-label pilot study. J Pain Symptom Manage. 2000;20(4):280–285. doi:10.1016/s0885-3924(00)00181-0
  • Rosenstock J, Tuchman M, LaMoreaux L, Sharma U. Pregabalin for the treatment of painful diabetic peripheral neuropathy: a double-blind, placebo-controlled trial. Pain. 2004;110(3):628–638. doi:10.1016/j.pain.2004.05.001
  • Sindrup SH, Otto M, Finnerup NB, Jensen TS. Antidepressants in the treatment of neuropathic pain. Basic Clin Pharmacol Toxicol. 2005;96(6):399–409. doi:10.1111/j.1742-7843.2005.pto_96696601.x
  • Max MB. Endogenous monoamine analgesic systems: amitriptyline in painful diabetic neuropathy. Anesth Prog. 1987;34(4):123–127.
  • Max MB, Lynch SA, Muir J, Shoaf SE, Smoller B, Dubner R. Effects of desipramine, amitriptyline, and fluoxetine on pain in diabetic neuropathy. N Engl J Med. 1992;326(19):1250–1256. doi:10.1056/NEJM199205073261904
  • Bansal D, Bhansali A, Hota D, Chakrabarti A, Dutta P. Amitriptyline vs. pregabalin in painful diabetic neuropathy: a randomized double blind clinical trial. Diabet Med. 2009;26(10):1019–1026. doi:10.1111/j.1464-5491.2009.02806.x
  • Cohen K, Shinkazh N, Frank J, Israel I, Fellner C. Pharmacological treatment of diabetic peripheral neuropathy. P T. 2015;40(6):372–388.
  • Vinik AI, Shapiro DY, Rauschkolb C, et al. A randomized withdrawal, placebo-controlled study evaluating the efficacy and tolerability of tapentadol extended release in patients with chronic painful diabetic peripheral neuropathy. Diabetes Care. 2014;37(8):2302–2309. doi:10.2337/dc13-2291
  • Blonde L, Umpierrez GE, Reddy SS, et al. American Association of Clinical Endocrinology Clinical Practice Guideline: developing a Diabetes Mellitus Comprehensive Care Plan-2022 Update. Endocr Pract. 2022;28(10):923–1049. doi:10.1016/j.eprac.2022.08.002
  • Morello CM, Leckband SG, Stoner CP, Moorhouse DF, Sahagian GA. Randomized double-blind study comparing the efficacy of gabapentin with amitriptyline on diabetic peripheral neuropathy pain. Arch Intern Med. 1999;159(16):1931–1937. doi:10.1001/archinte.159.16.1931
  • American Diabetes Association. Chapter 11. Microvascular Complications and Foot Care: standards of Medical Care in Diabetes—2021. Diabetes Care. 2020;44(Supplement_1):S151–S167. doi:10.2337/dc21-S011
  • Argoff CE, Galer BS, Jensen MP, Oleka N, Gammaitoni AR. Effectiveness of the lidocaine patch 5% on pain qualities in three chronic pain states: assessment with the Neuropathic Pain Scale. Curr Med Res Opin. 2004;20(Suppl 2):S21–S28. doi:10.1185/030079904X12960
  • Meier T, Wasner G, Faust M, et al. Efficacy of lidocaine patch 5% in the treatment of focal peripheral neuropathic pain syndromes: a randomized, double-blind, placebo-controlled study. Pain. 2003;106(1–2):151–158. doi:10.1016/s0304-3959(03)00317-8
  • Barbano RL, Herrmann DN, Hart-Gouleau S, Pennella-Vaughan J, Lodewick PA, Dworkin RH. Effectiveness, tolerability, and impact on quality of life of the 5% lidocaine patch in diabetic polyneuropathy. Arch Neurol. 2004;61(6):914–918. doi:10.1001/archneur.61.6.914
  • Baron R, Mayoral V, Leijon G, Binder A, Steigerwald I, Serpell M. Efficacy and safety of 5% lidocaine (lignocaine) medicated plaster in comparison with pregabalin in patients with postherpetic neuralgia and diabetic polyneuropathy: interim analysis from an open-label, two-stage adaptive, randomized, controlled trial. Clin Drug Investig. 2009;29(4):231–241. doi:10.2165/00044011-200929040-00002
  • Baron R, Mayoral V, Leijon G, Binder A, Steigerwald I, Serpell M. 5% lidocaine medicated plaster versus pregabalin in post-herpetic neuralgia and diabetic polyneuropathy: an open-label, non-inferiority two-stage RCT study. Curr Med Res Opin. 2009;25(7):1663–1676. doi:10.1185/03007990903047880
  • Baron R, Mayoral V, Leijon G, Binder A, Steigerwald I, Serpell M. 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;25(7):1677–1687. doi:10.1185/03007990903048078
  • Anand P, Privitera R, Donatien P, et al. Reversing painful and non-painful diabetic neuropathy with the capsaicin 8% patch: clinical evidence for pain relief and restoration of function via nerve fiber regeneration. Front Neurol. 2022;13:998904. doi:10.3389/fneur.2022.998904
  • Simpson DM, Robinson-Papp J, Van J, et al. Capsaicin 8% Patch in Painful Diabetic Peripheral Neuropathy: a Randomized, Double-Blind, Placebo-Controlled Study. J Pain. 2017;18(1):42–53. doi:10.1016/j.jpain.2016.09.008
  • Capsaicin Study Group. Effect of treatment with capsaicin on daily activities of patients with painful diabetic neuropathy. Diabetes Care. 1992;15(2):159–165. doi:10.2337/diacare.15.2.159
  • The Capsaicin Study Group. Treatment of painful diabetic neuropathy with topical capsaicin. A multicenter, double-blind, vehicle-controlled study. Arch Intern Med. 1991;151(11):2225–2229. doi:10.1001/archinte.151.11.2225
  • Tandan R, Lewis GA, Krusinski PB, Badger GB, Fries TJ. Topical capsaicin in painful diabetic neuropathy. Controlled study with long-term follow-up. Diabetes Care. 1992;15(1):8–14. doi:10.2337/diacare.15.1.8
  • Biesbroeck R, Bril V, Hollander P, et al. A double-blind comparison of topical capsaicin and oral amitriptyline in painful diabetic neuropathy. Adv Ther. 1995;12(2):111–120.
  • Mou J, Paillard F, Turnbull B, Trudeau J, Stoker M, Katz NP. Efficacy of Qutenza® (capsaicin) 8% patch for neuropathic pain: a meta-analysis of the Qutenza Clinical Trials Database. Pain. 2013;154(9):1632–1639. doi:10.1016/j.pain.2013.04.044
  • Ho KY, Huh BK, White WD, Yeh CC, Miller EJ. Topical amitriptyline versus lidocaine in the treatment of neuropathic pain. Clin J Pain. 2008;24(1):51–55. doi:10.1097/AJP.0b013e318156db26
  • Kiani J, Ahmad Nasrollahi S, Esna-Ashari F, Fallah P, Sajedi F. Amitriptyline 2% cream vs. capsaicin 0.75% cream in the treatment of painful diabetic neuropathy (Double blind, randomized clinical trial of efficacy and safety). Iran J Pharm Res. 2015;14(4):1263–1268.
  • de Vos CC, Meier K, Zaalberg PB, et al. Spinal cord stimulation in patients with painful diabetic neuropathy: a multicentre randomized clinical trial. Pain®. 2014;155(11):2426–2431. doi:10.1016/j.pain.2014.08.031
  • Slangen R, Schaper NC, Faber CG, et al. Spinal cord stimulation and pain relief in painful diabetic peripheral neuropathy: a prospective two-center randomized controlled trial. Dia Care. 2014;37(11):3016–3024. doi:10.2337/dc14-0684
  • de Vos CC, Bom MJ, Vanneste S, Lenders MWPM, de Ridder D. Burst spinal cord stimulation evaluated in patients with failed back surgery syndrome and painful diabetic neuropathy. Neuromodulation. 2014;17(2):152–159. doi:10.1111/ner.12116
  • van Beek M, Geurts JW, Slangen R, et al. Severity of Neuropathy Is Associated With Long-term Spinal Cord Stimulation Outcome in Painful Diabetic Peripheral Neuropathy: five-Year Follow-up of a Prospective Two-Center Clinical Trial. Diabetes Care. 2018;41(1):32–38. doi:10.2337/dc17-0983
  • Petersen EA, Stauss TG, Scowcroft JA, et al. Effect of High-frequency (10-kHz) Spinal Cord Stimulation in Patients With Painful Diabetic Neuropathy: a Randomized Clinical Trial. JAMA Neurol. 2021;78(6):687–698. doi:10.1001/jamaneurol.2021.0538
  • Petersen EA, Stauss TG, Scowcroft JA, et al. High-Frequency 10-kHz Spinal Cord Stimulation Improves Health-Related Quality of Life in Patients With Refractory Painful Diabetic Neuropathy: 12-Month Results From a Randomized Controlled Trial. Mayo Clin Proc Innov Qual Outcomes. 2022;6(4):347–360. doi:10.1016/j.mayocpiqo.2022.05.003
  • Petersen EA, Stauss TG, Scowcroft JA, et al. Long-term efficacy of high-frequency (10 kHz) spinal cord stimulation for the treatment of painful diabetic neuropathy: 24-Month results of a randomized controlled trial. Diabet Res Clin Pract. 2023;203:110865. doi:10.1016/j.diabres.2023.110865
  • D’Souza RS, ElSaban M, Martinez Alvarez GA, Jin MY, Kubrova E, Hassett LC. Treatment of pain in length-dependent peripheral neuropathy with the use of spinal cord stimulation: a systematic review. Pain Med. 2023;24(Supplement_2):S24–S32. doi:10.1093/pm/pnad091
  • Deer T, Pope J, Benyamin R, et al. Prospective, Multicenter, Randomized, Double-Blinded, Partial Crossover Study to Assess the Safety and Efficacy of the Novel Neuromodulation System in the Treatment of Patients With Chronic Pain of Peripheral Nerve Origin. Neuromodulation. 2016;19(1):91–100. doi:10.1111/ner.12381
  • Chmiela MA, Hendrickson M, Hale J, et al. Direct Peripheral Nerve Stimulation for the Treatment of Complex Regional Pain Syndrome: a 30-Year Review. Neuromodulation. 2021;24(6):971–982. doi:10.1111/ner.13295
  • Gilmore C, Ilfeld B, Rosenow J, et al. Percutaneous peripheral nerve stimulation for the treatment of chronic neuropathic postamputation pain: a multicenter, randomized, placebo-controlled trial. Reg Anesth Pain Med. 2019;44(6):637–645. doi:10.1136/rapm-2018-100109
  • Gilligan C, Volschenk W, Russo M, et al. Three-Year Durability of Restorative Neurostimulation Effectiveness in Patients With Chronic Low Back Pain and Multifidus Muscle Dysfunction. Neuromodulation. 2023;26(1):98–108. doi:10.1016/j.neurom.2022.08.457
  • Gilmore CA, Kapural L, McGee MJ, Boggs JW. Percutaneous Peripheral Nerve Stimulation (PNS) for the Treatment of Chronic Low Back Pain Provides Sustained Relief. Neuromodulation. 2019;22(5):615–620. doi:10.1111/ner.12854
  • Ilfeld BM, Plunkett A, Vijjeswarapu AM, et al. Percutaneous Peripheral Nerve Stimulation (Neuromodulation) for Postoperative Pain: a Randomized, Sham-controlled Pilot Study. Anesthesiology. 2021;135(1):95–110. doi:10.1097/ALN.0000000000003776
  • Narouze S, Benzon HT, Provenzano D, et al. Interventional Spine and Pain Procedures in Patients on Antiplatelet and Anticoagulant Medications (Second Edition): guidelines From the American Society of Regional Anesthesia and Pain Medicine, the European Society of Regional Anaesthesia and Pain Therapy, the American Academy of Pain Medicine, the International Neuromodulation Society, the North American Neuromodulation Society, and the World Institute of Pain. Reg Anesth Pain Med. 2018;43(3):225–262. doi:10.1097/AAP.0000000000000700
  • Strand N, D’Souza RS, Hagedorn JM, et al. Evidence-Based Clinical Guidelines from the American Society of Pain and Neuroscience for the Use of Implantable Peripheral Nerve Stimulation in the Treatment of Chronic Pain. J Pain Res. 2022;15:2483–2504. doi:10.2147/JPR.S362204
  • Hanyu-Deutmeyer A, Pritzlaff SG. Peripheral Nerve Stimulation for the 21st Century: sural, Superficial Peroneal, and Tibial Nerves. Pain Med. 2020;21(Suppl 1):S64–S67. doi:10.1093/pm/pnaa202
  • Eldabe S, Espinet A, Wahlstedt A, et al. Retrospective Case Series on the Treatment of Painful Diabetic Peripheral Neuropathy With Dorsal Root Ganglion Stimulation. Neuromodulation. 2018;21(8):787–792. doi:10.1111/ner.12767
  • Dellon AL. Treatment of symptomatic diabetic neuropathy by surgical decompression of multiple peripheral nerves. Plast Reconstr Surg. 1992;89(4):689–697.
  • Aszmann OC, Kress KM, Dellon AL. Results of decompression of peripheral nerves in diabetics: a prospective, blinded study. Plast Reconstr Surg. 2000;106(4):816–822. doi:10.1097/00006534-200009040-00010
  • D’Souza RS, Langford B, Dombovy-Johnson M, Abd-Elsayed A. Neuromodulation Interventions for the Treatment of Painful Diabetic Neuropathy: a Systematic Review. Curr Pain Headache Rep. 2022;26(5):365–377. doi:10.1007/s11916-022-01035-9
  • Hamza MA, White PF, Craig WF, et al. Percutaneous electrical nerve stimulation: a novel analgesic therapy for diabetic neuropathic pain. Diabetes Care. 2000;23(3):365–370. doi:10.2337/diacare.23.3.365
  • Dabby R, Sadeh M, Goldberg I, Finkelshtein V. Electrical stimulation of the posterior tibial nerve reduces neuropathic pain in patients with polyneuropathy. JPR. 2017;10:2717–2723. doi:10.2147/JPR.S137420
  • Sokal P, Harat M, Zieliński P, Kieronska S. Tibial nerve stimulation with a miniature, wireless stimulator in chronic peripheral neuropathic pain. JPR. 2017;10:613–619. doi:10.2147/JPR.S128861
  • Franken G, Debets J, Joosten EAJ. Nonlinear Relation Between Burst Dorsal Root Ganglion Stimulation Amplitude and Behavioral Outcome in an Experimental Model of Painful Diabetic Peripheral Neuropathy. Neuromodulation: journal of the International Neuromodulation. Society. 2020;23(2):158–166. doi:10.1111/ner.13070
  • Franken G, Douven P, Debets J, Joosten EAJ. Conventional Dorsal Root Ganglion Stimulation in an Experimental Model of Painful Diabetic Peripheral Neuropathy: a Quantitative Immunocytochemical Analysis of Intracellular γ-Aminobutyric Acid in Dorsal Root Ganglion Neurons. Neuromodulation. 2021;24(4):639–645. doi:10.1111/ner.13398
  • Chapman KB, Van Roosendaal BKW, Van Helmond N, Yousef TA. Unilateral Dorsal Root Ganglion Stimulation Lead Placement With Resolution of Bilateral Lower Extremity Symptoms in Diabetic Peripheral Neuropathy. Cureus. 2020;12(9):e10735. doi:10.7759/cureus.10735
  • Groenen PS, Van Helmond N, Chapman KB, et al. Chemotherapy-Induced Peripheral Neuropathy Treated with Dorsal Root Ganglion Stimulation. Pain Med. 2019;20(4):857–859. doi:10.1093/pm/pny209
  • Maino P, Koetsier E, Kaelin-Lang A, Gobbi C, Perez R. Efficacious Dorsal Root Ganglion Stimulation for Painful Small Fiber Neuropathy: a Case Report. Pain Physician. 2017;20(3):E459–E463.
  • Grabnar M, Kim C. Dorsal Root Ganglion Stimulation for Treatment of Chemotherapy-Induced Neuropathy. Am J Phys Med Rehabilitation. 2020;1(4):2020–2022. doi:10.1097/phm.0000000000001542
  • Karri J, Bruel B. Dorsal Root Ganglion Stimulation for Post-Lyme Disease Chronic Peripheral Neuropathic Pain. Neuromodulation. 2020. doi:10.1111/ner.13136
  • Falowski S, Pope JE, Raza A. Early US Experience With Stimulation of the Dorsal Root Ganglia for the Treatment of Peripheral Neuropathy in the Lower Extremities: a Multicenter Retrospective Case Series. Neuromodulation. 2019;22(1):96–100. doi:10.1111/ner.12860
  • Pope JE, Deer TR, Amirdelfan K, McRoberts WP, Azeem N. The Pharmacology of Spinal Opioids and Ziconotide for the Treatment of Non-Cancer Pain. Curr Neuropharmacol. 2017;15(2):206–216. doi:10.2174/1570159x14666160210142339
  • Pope JE, Deer TR. Ziconotide: a clinical update and pharmacologic review. Expert Opin Pharmacother. 2013;14(7):957–966. doi:10.1517/14656566.2013.784269
  • Deer TR, Pope JE, Hayek SM, et al. The Polyanalgesic Consensus Conference (PACC): recommendations on Intrathecal Drug Infusion Systems Best Practices and Guidelines. Neuromodulation. 2017;20(2):96–132. doi:10.1111/ner.12538
  • Smith HS, Deer TR. Safety and efficacy of intrathecal ziconotide in the management of severe chronic pain. Ther Clin Risk Manag. 2009;5(3):521–534. doi:10.2147/tcrm.s4438
  • Staats PS, Yearwood T, Charapata SG, et al. Intrathecal ziconotide in the treatment of refractory pain in patients with cancer or AIDS: a randomized controlled trial. JAMA. 2004;291(1):63–70. doi:10.1001/jama.291.1.63
  • Wallace MS. Ziconotide: a new nonopioid intrathecal analgesic for the treatment of chronic pain. Expert Rev Neurother. 2006;6(10):1423–1428. doi:10.1586/14737175.6.10.1423
  • Rauck RL, Wallace MS, Leong MS, et al. A randomized, double-blind, placebo-controlled study of intrathecal ziconotide in adults with severe chronic pain. J Pain Symptom Manage. 2006;31(5):393–406. doi:10.1016/j.jpainsymman.2005.10.003
  • Shao MM, Khazen O, Hellman A, et al. Effect of First-Line Ziconotide Intrathecal Drug Therapy for Neuropathic Pain on Disability, Emotional Well-Being, and Pain Catastrophizing. World Neurosurg. 2021;145:e340–e347. doi:10.1016/j.wneu.2020.10.079
  • Mendez CE, Wainaina N, Walker RJ, et al. Preoperative Diabetes Optimization Program. Clin Diabetes. 2018;36(1):68–71. doi:10.2337/cd17-0088
  • Underwood P, Askari R, Hurwitz S, Chamarthi B, Garg R. Preoperative A1C and clinical outcomes in patients with diabetes undergoing major noncardiac surgical procedures. Diabetes Care. 2014;37(3):611–616. doi:10.2337/dc13-1929
  • Deer TR, Russo MA, Grider JS, et al. The Neurostimulation Appropriateness Consensus Committee (NACC): recommendations for Surgical Technique for Spinal Cord Stimulation. Neuromodulation. 2022;25(1):1–34. doi:10.1016/j.neurom.2021.10.015
  • Goel V, Kumar V, Agrawal SN, et al. Outcomes Associated With Infection of Chronic Pain Spinal Implantable Electronic Devices: insights From a Nationwide Inpatient Sample Study. Neuromodulation. 2021;24(1):126–134. doi:10.1111/ner.13263
  • Falowski SM, Provenzano DA, Xia Y, Doth AH. Spinal Cord Stimulation Infection Rate and Risk Factors: results From a United States Payer Database. Neuromodulation. 2019;22(2):179–189. doi:10.1111/ner.12843
  • Hoelzer BC, Bendel MA, Deer TR, et al. Spinal Cord Stimulator Implant Infection Rates and Risk Factors: a Multicenter Retrospective Study. Neuromodulation. 2017;20(6):558–562. doi:10.1111/ner.12609
  • Stewart PS, Bjarnsholt T. Risk factors for chronic biofilm-related infection associated with implanted medical devices. Clin Microbiol Infect. 2020;26(8):1034–1038. doi:10.1016/j.cmi.2020.02.027
  • Gabriel RA, Hylton DJ, Burton BN, Schmidt H, Waterman RS. The association of preoperative haemoglobin A1c with 30-day postoperative surgical site infection following non-cardiac surgery. J Perioper Pract. 2019;30(10):320–325. doi:10.1177/1750458919886183
  • Sethuraman RM, Parida S, Sethuramachandran A, Selvam P. A1C as a Prognosticator of Perioperative Complications of Diabetes: a Narrative Review. Turk J Anaesthesiol Reanim. 2022;50(2):79–85. doi:10.5152/TJAR.2021.854
  • Walid MS, Newman BF, Yelverton JC, Nutter JP, Ajjan M, Robinson JSJ. Prevalence of previously unknown elevation of glycosylated hemoglobin in spine surgery patients and impact on length of stay and total cost. J Hosp Med. 2010;5(1):E10–14. doi:10.1002/jhm.541
  • Adams AL, Paxton EW, Wang JQ, et al. Surgical outcomes of total knee replacement according to diabetes status and glycemic control, 2001 to 2009. J Bone Joint Surg Am. 2013;95(6):481–487. doi:10.2106/JBJS.L.00109
  • Bock M, Johansson T, Fritsch G, et al. The impact of preoperative testing for blood glucose concentration and haemoglobin A1c on mortality, changes in management and complications in noncardiac elective surgery: a systematic review. Eur J Anaesthesiol. 2015;32(3):152–159. doi:10.1097/EJA.0000000000000117
  • Maradit Kremers H, Lewallen LW, Mabry TM, Berry DJ, Berbari EF, Osmon DR. Diabetes mellitus, hyperglycemia, hemoglobin A1C and the risk of prosthetic joint infections in total Hip and knee arthroplasty. J Arthroplasty. 2015;30(3):439–443. doi:10.1016/j.arth.2014.10.009
  • Geddes LA. History of magnetic stimulation of the nervous system. J Clin Neurophysiol. 1991;8(1):3–9. doi:10.1097/00004691-199101000-00003
  • Malmivuo J. Magnetic Stimulation of Neural Tissue. In: Malmivuo J, Plonsey R editors. Bioelectromagnetism: Principles and Applications of Bioelectric and Biomagnetic Fields. Oxford University Press; 1995:45. doi:10.1093/acprof:oso/9780195058239.003.0022
  • Polson MJ, Barker AT, Freeston IL. Stimulation of nerve trunks with time-varying magnetic fields. Med Biol Eng Comput. 1982;20(2):243–244. doi:10.1007/BF02441362
  • Bedder M, Parker L Magnetic Peripheral Nerve Stimulation (mPNS) for Chronic Pain. Presented at: Fourth Annual ASPN Conference; 2022; Miami Beach. https://aspnpain.com/aspn-annual-conference-2022/.
  • Kapural L, Rosenberg J, Li S Safety and Efficacy of Axon Therapy (SEAT Study). Presented at: Fifth Annual ASPN Conference; 2023; Miami Beach.
  • Zhou M, Zhang Q, Huo M, et al. The mechanistic basis for the effects of electroacupuncture on neuropathic pain within the central nervous system. Biomed. Pharmacother. 2023;161:114516. doi:10.1016/j.biopha.2023.114516
  • Dimitrova A, Murchison C, Oken B. Acupuncture for the Treatment of Peripheral Neuropathy: a Systematic Review and Meta-Analysis. J Altern Complement Med. 2017;23(3):164–179. doi:10.1089/acm.2016.0155
  • Dietzel J, Habermann IV, Horder S, et al. Acupuncture in Patients with Diabetic Peripheral Neuropathy-Related Complaints: a Randomized Controlled Clinical Trial. J Clin Med. 2023;12(6):2103. doi:10.3390/jcm12062103
  • Feng Z, Cui S, Yang H, et al. Acupuncture for neuropathic pain: a meta-analysis of randomized control trials. Front Neurol. 2023:13. doi:10.3389/fneur.2022.1076993
  • Shin KM, Lee S, Lee EY, et al. Electroacupuncture for Painful Diabetic Peripheral Neuropathy: a Multicenter, Randomized, Assessor-Blinded, Controlled Trial. Diabetes Care. 2018;41(10):e141–e142. doi:10.2337/dc18-1254
  • Abuaisha BB, Costanzi JB, Boulton AJ. Acupuncture for the treatment of chronic painful peripheral diabetic neuropathy: a long-term study. Diabet Res Clin Pract. 1998;39(2):115–121. doi:10.1016/s0168-8227(97)00123-x
  • Garrow AP, Xing M, Vere J, Verrall B, Wang L, Jude EB. Role of acupuncture in the management of diabetic painful neuropathy (DPN): a pilot RCT. Acupunct Med. 2014;32(3):242–249. doi:10.1136/acupmed-2013-010495
  • Chao MT, Schillinger D, Nguyen U, et al. A Randomized Clinical Trial of Group Acupuncture for Painful Diabetic Neuropathy Among Diverse Safety Net Patients. Pain Med. 2019;20(11):2292–2302. doi:10.1093/pm/pnz117
  • Meyer-Hamme G, Friedemann T, Greten J, Gerloff C, Schroeder S. Electrophysiologically verified effects of acupuncture on diabetic peripheral neuropathy in type 2 diabetes: the randomized, partially double-blinded, controlled ACUDIN trial. J Diabetes. 2021;13(6):469–481. doi:10.1111/1753-0407.13130
  • Zheng H-T, Li Y-F, Yuan S-X, Zhang C-G, Chen G-M, Zhang L-F. Observations on 52 patients with diabetic peripheral neuropathy treated by needling combined with drug. J Acupuncture Tuina Sci. 2004;2(6):24–26. doi:10.1007/BF02848393
  • Hassanien M, Elawamy A, Kamel EZ, et al. Perineural Platelet-Rich Plasma for Diabetic Neuropathic Pain, Could It Make a Difference? Pain Med. 2020;21(4):757–765. doi:10.1093/pm/pnz140
  • Xu L, Sun Z, Casserly E, Nasr C, Cheng J, Xu J. Advances in Interventional Therapies for Painful Diabetic Neuropathy: a Systematic Review. Anesth Analg. 2022;134(6):1215–1228. doi:10.1213/ANE.0000000000005860
  • Cheng J, Daftari A, Zhou L. Sympathetic blocks provided sustained pain relief in a patient with refractory painful diabetic neuropathy. Case Rep Anesthesiol. 2012;2012:285328. doi:10.1155/2012/285328
  • Sun H, He M, Pang J, Guo X, Huo Y, Ma J. Continuous Lumbar Sympathetic Blockade Enhances the Effect of Lumbar Sympatholysis on Refractory Diabetic Neuropathy: a Randomized Controlled Trial. Diabetes Ther. 2020;11(11):2647–2655. doi:10.1007/s13300-020-00918-7
  • Ding Y, Yao P, Li H, Zhao R, Zhao G. Evaluation of combined radiofrequency and chemical blockade of multi-segmental lumbar sympathetic ganglia in painful diabetic peripheral neuropathy. J Pain Res. 2018;11:1375–1382. doi:10.2147/JPR.S175514
  • Salehi H, Moussaei M, Kamiab Z, Vakilian A. The effects of botulinum toxin type A injection on pain symptoms, quality of life, and sleep quality of patients with diabetic neuropathy: a randomized double-blind clinical trial. Iran J Neurol. 2019;18(3):99–107.
  • Yuan RY, Sheu JJ, Yu JM, et al. Botulinum toxin for diabetic neuropathic pain: a randomized double-blind crossover trial. Neurology. 2009;72(17):1473–1478. doi:10.1212/01.wnl.0000345968.05959.cf
  • Ghasemi M, Ansari M, Basiri K, Shaigannejad V. The effects of intradermal botulinum toxin type a injections on pain symptoms of patients with diabetic neuropathy. J Res Med Sci. 2014;19(2):106–111.
  • Lakhan SE, Velasco DN, Tepper D. Botulinum Toxin-A for Painful Diabetic Neuropathy: a Meta-Analysis. Pain Med. 2015;16(9):1773–1780. doi:10.1111/pme.12728
  • Zaheer A, Zaheer F, Saeed HY, Tahir Z, Tahir MW. A Review of Alternative Treatment Options in Diabetic Polyneuropathy. Cureus. 2021;13(4). doi:10.7759/cureus.14600
  • 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(11):2365–2370. doi:10.2337/dc06-1216
  • Ziegler D, Low PA, Litchy WJ, et al. Efficacy and safety of antioxidant treatment with α-lipoic acid over 4 years in diabetic polyneuropathy: the NATHAN 1 trial. Diabetes Care. 2011;34(9):2054–2060. doi:10.2337/dc11-0503
  • Garcia-Alcala H, Santos Vichido CI, Islas Macedo S, et al. Treatment with α-Lipoic Acid over 16 Weeks in Type 2 Diabetic Patients with Symptomatic Polyneuropathy Who Responded to Initial 4-Week High-Dose Loading. J Diabetes Res. 2015;2015:189857. doi:10.1155/2015/189857
  • Chiechio S, Copani A, Gereau RW 4th, Nicoletti F. Acetyl-L-carnitine in neuropathic pain: experimental data. CNS Drugs. 2007;21 Suppl 1:31–38. doi:10.2165/00023210-200721001-00005
  • De Grandis D, Minardi C. Acetyl-L-carnitine (levacecarnine) in the treatment of diabetic neuropathy. A long-term, randomised, double-blind, placebo-controlled study. Drugs R D. 2002;3(4):223–231. doi:10.2165/00126839-200203040-00001
  • Sima AAF, Calvani M, Mehra M, Amato A. 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(1):89–94. doi:10.2337/diacare.28.1.89
  • Baltrusch S. The Role of Neurotropic B Vitamins in Nerve Regeneration. Biomed Res Int. 2021;2021:9968228. doi:10.1155/2021/9968228
  • Feldman EL, Callaghan BC, Pop-Busui R, et al. Diabetic neuropathy. Nat Rev Dis Primers. 2019;5(1):41. doi:10.1038/s41572-019-0092-1
  • Ismail-Beigi F, Craven T, Banerji MA, et al. Effect of intensive treatment of hyperglycaemia on microvascular outcomes in type 2 diabetes: an analysis of the ACCORD randomised trial. Lancet. 2010;376(9739):419–430. doi:10.1016/S0140-6736(10)60576-4
  • Buse JB, Bigger JT, Byington RP, et al. Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial: design and methods. Am J Cardiol. 2007;99(12A):21i–33i. doi:10.1016/j.amjcard.2007.03.003
  • Qaseem A. Hemoglobin A1c Targets for Glycemic Control With Pharmacologic Therapy for Nonpregnant Adults With Type 2 Diabetes Mellitus: a Guidance Statement Update From the American College of Physicians. Int J Med. 2018:168:58. doi:10.7326/M17-0939
  • Charles M, Ejskjaer N, Witte DR, Borch-Johnsen K, Lauritzen T, Sandbaek A. Prevalence of neuropathy and peripheral arterial disease and the impact of treatment in people with screen-detected type 2 diabetes: the ADDITION-Denmark study. Diabetes Care. 2011;34(10):2244–2249. doi:10.2337/dc11-0903
  • Nathan DM, Genuth S, Lachin J, et al. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med. 1993;329(14):977–986. doi:10.1056/NEJM199309303291401
  • Duckworth W, Abraira C, Moritz T, et al. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med. 2009;360(2):129–139. doi:10.1056/NEJMoa0808431
  • Singleton JR, Marcus RL, Jackson JE, Lessard K, Graham T, Smith TE. Exercise increases cutaneous nerve density in diabetic patients without neuropathy. Ann Clin Transl Neurol. 2014;1(10):844–849. doi:10.1002/acn3.125
  • Singleton JR, Marcus RL, Lessard MK, Jackson JE, Smith AG. Supervised exercise improves cutaneous reinnervation capacity in metabolic syndrome patients. Ann Neurol. 2015;77(1):146–153. doi:10.1002/ana.24310
  • van Laake-Geelen CCM, Smeets RJEM, Quadflieg SPAB, Kleijnen J, Verbunt JA. The effect of exercise therapy combined with psychological therapy on physical activity and quality of life in patients with painful diabetic neuropathy: a systematic review. Scand J Pain. 2019;19(3):433–439. doi:10.1515/sjpain-2019-0001
  • Vileikyte L, Leventhal H, Gonzalez JS, et al. Diabetic peripheral neuropathy and depressive symptoms: the association revisited. Diabetes Care. 2005;28(10):2378–2383. doi:10.2337/diacare.28.10.2378
  • Yoshida S, Hirai M, Suzuki S, Awata S, Oka Y. Neuropathy is associated with depression independently of health-related quality of life in Japanese patients with diabetes. Psychiatry Clin Neurosci. 2009;63(1):65–72. doi:10.1111/j.1440-1819.2008.01889.x
  • Gore M, Brandenburg NA, Dukes E, Hoffman DL, Tai KS, Stacey B. Pain severity in diabetic peripheral neuropathy is associated with patient functioning, symptom levels of anxiety and depression, and sleep. J Pain Symptom Manage. 2005;30(4):374–385. doi:10.1016/j.jpainsymman.2005.04.009
  • Jain R, Jain S, Raison CL, Maletic V. Painful diabetic neuropathy is more than pain alone: examining the role of anxiety and depression as mediators and complicators. Curr Diab Rep. 2011;11(4):275–284. doi:10.1007/s11892-011-0202-2
  • Sullivan MJL, Lynch ME, Clark AJ. Dimensions of catastrophic thinking associated with pain experience and disability in patients with neuropathic pain conditions. Pain. 2005;113(3):310–315. doi:10.1016/j.pain.2004.11.003
  • Geelen CC, Kindermans HP, van den Bergh JP, Verbunt JA. Perceived Physical Activity Decline as a Mediator in the Relationship Between Pain Catastrophizing, Disability, and Quality of Life in Patients with Painful Diabetic Neuropathy. Pain Pract. 2017;17(3):320–328. doi:10.1111/papr.12449
  • Benbow SJ, Wallymahmed ME, MacFarlane IA. Diabetic peripheral neuropathy and quality of life. QJM. 1998;91(11):733–737. doi:10.1093/qjmed/91.11.733
  • Galer BS, Gianas A, Jensen MP. Painful diabetic polyneuropathy: epidemiology, pain description, and quality of life. Diabet Res Clin Pract. 2000;47(2):123–128. doi:10.1016/s0168-8227(99)00112-6
  • Teixeira E. The effect of mindfulness meditation on painful diabetic peripheral neuropathy in adults older than 50 years. Holist Nurs Pract. 2010;24(5):277–283. doi:10.1097/HNP.0b013e3181f1add2
  • Otis JD, Sanderson K, Hardway C, Pincus M, Tun C, Soumekh S. A randomized controlled pilot study of a cognitive-behavioral therapy approach for painful diabetic peripheral neuropathy. J Pain. 2013;14(5):475–482. doi:10.1016/j.jpain.2012.12.013
  • Taheri AA, Foroughi AA, Mohammadian Y, et al. The Effectiveness of Acceptance and Commitment Therapy on Pain Acceptance and Pain Perception in Patients with Painful Diabetic Neuropathy: a Randomized Controlled Trial. Diabetes Ther. 2020;11(8):1695–1708. doi:10.1007/s13300-020-00851-9
  • Higgins DM, Heapy AA, Buta E, et al. A randomized controlled trial of cognitive behavioral therapy compared with diabetes education for diabetic peripheral neuropathic pain. J Health Psychol. 2022;27(3):649–662. doi:10.1177/1359105320962262
  • Izgu N, Gok Metin Z, Karadas C, Ozdemir L, Metinarikan N, Corapcıoglu D. Progressive Muscle Relaxation and Mindfulness Meditation on Neuropathic Pain, Fatigue, and Quality of Life in Patients With Type 2 Diabetes: a Randomized Clinical Trial. J Nurs Scholarsh. 2020;52(5):476–487. doi:10.1111/jnu.12580
  • Nathan HJ, Poulin P, Wozny D, et al. Randomized Trial of the Effect of Mindfulness-Based Stress Reduction on Pain-Related Disability, Pain Intensity, Health-Related Quality of Life, and A1C in Patients With Painful Diabetic Peripheral Neuropathy. Clin Diabetes. 2017;35(5):294–304. doi:10.2337/cd17-0077
  • Hussain N, Said ASA. Mindfulness-Based Meditation Versus Progressive Relaxation Meditation: impact on Chronic Pain in Older Female Patients With Diabetic Neuropathy. J Evid Based Integr Med. 2019;24:2515690X19876599. doi:10.1177/2515690X19876599
  • Eccleston C, Hearn L, Williams AC. Psychological therapies for the management of chronic neuropathic pain in adults. Cochrane Database Syst Rev. 2015;2015(10):CD011259. doi:10.1002/14651858.CD011259.pub2
  • Castelnuovo G, Giusti EM, Manzoni GM, et al. Psychological Treatments and Psychotherapies in the Neurorehabilitation of Pain: evidences and Recommendations from the Italian Consensus Conference on Pain in Neurorehabilitation. Front Psychol. 2016;7:115. doi:10.3389/fpsyg.2016.00115
  • Bai Y, Ma JH, Yu Y, Wang ZW. Effect of Cognitive-Behavioral Therapy or Mindfulness Therapy on Pain and Quality of Life in Patients with Diabetic Neuropathy: a Systematic Review and Meta-Analysis. Pain Manag Nurs. 2022;23(6):861–870. doi:10.1016/j.pmn.2022.05.005
  • Veves A, Backonja M, Malik RA. Painful diabetic neuropathy: epidemiology, natural history, early diagnosis, and treatment options. Pain Med. 2008;9(6):660–674. doi:10.1111/j.1526-4637.2007.00347.x
  • Callaghan BC, Little AA, Feldman EL, Hughes RAC. Enhanced glucose control for preventing and treating diabetic neuropathy. Cochrane Database Syst Rev. 2012;6(6):CD007543. doi:10.1002/14651858.CD007543.pub2
  • Balducci S, Iacobellis G, Parisi L, et al. Exercise training can modify the natural history of diabetic peripheral neuropathy. J Diabetes Complications. 2006;20(4):216–223. doi:10.1016/j.jdiacomp.2005.07.005
  • Holmes CJ, Hastings MK. The Application of Exercise Training for Diabetic Peripheral Neuropathy. J Clin Med. 2021;10(21). doi:10.3390/jcm10215042
  • Chatchawan U, Eungpinichpong W, Plandee P, Yamauchi J. Effects of Thai foot massage on balance performance in diabetic patients with peripheral neuropathy: a randomized parallel-controlled trial. Med Sci Monit Basic Res. 2015;21:68–75. doi:10.12659/MSMBR.894163
  • Busse JW, Wang L, Kamaleldin M, et al. Opioids for Chronic Noncancer Pain: a Systematic Review and Meta-analysis. JAMA. 2018;320(23):2448–2460. doi:10.1001/jama.2018.18472
  • 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(20):1758–1765. doi:10.1212/WNL.0b013e3182166ebe
  • Liampas A, Rekatsina M, Vadalouca A, Paladini A, Varrassi G, Zis P. Non-Pharmacological Management of Painful Peripheral Neuropathies: a Systematic Review. Adv Ther. 2020;37(10):4096–4106. doi:10.1007/s12325-020-01462-3