Rare Diseases Report 2022

The broad and challenging – but promising – landscape of peripheral neuropathy


 

References

1. Barrell K, Smith AG. Peripheral neuropathy. Med Clin North Am. 2019 Mar;103(2):383-97. doi: 10.1016/j.mcna.2018.10.006.

2. Selvarajah D et al. Diabetic peripheral neuropathy: Advances in diagnosis and strategies for screening and early intervention. Lancet Diabetes Endocrinol. 2019 Dec;7(12):938-48. doi: 10.1016/S2213-8587(19)30081-6.

3. Snyder MJ et al. Treating painful diabetic peripheral neuropathy: An update. Am Fam Physician. 2016 Aug;94(3):227-334.

4. Sharma S et al. Assessment of diabetic neuropathy using a point-of-care nerve conduction device shows significant associations with the LDIFLARE method and clinical neuropathy scoring. J Diabetes Sci Technol. 2014 Jan;9(1):123-31. doi: 10.1177/1932296814551044.

5. Zografou I et al. Validation of Neuropad in the assessment of peripheral diabetic neuropathy in patients with diabetes mellitus versus the Michigan Neuropathy Screening Instrument, 10g monofilament application and biothesiometer measurement. Curr Vasc Pharmacol. 2020;18(5):517-22. doi: 10.2174/1570161117666190723155324.

6. Tentolouris N et al. Moisture status of the skin of the feet assessed by the visual test Neuropad correlates with foot ulceration in diabetes. Diabetes Care. 2010;33(5):1112-4. doi: 10.2337/dc09-2027.

7. Mao F et al. Sudoscan is an effective screening method for asymptomatic diabetic neuropathy in Chinese type 2 diabetes mellitus patients. J Diabetes Investig. 2017 May;8(3):363-8. doi: 10.1111/jdi.12575.

8. Kalteniece A et al. Corneal confocal microscopy is a rapid reproducible ophthalmic technique for quantifying corneal nerve abnormalities. PLoS One. 2017 Aug;12(8):e0183040. doi: 10.1371/journal.pone.0183040.

9. Gad H et al. Corneal confocal microscopy for the diagnosis of diabetic peripheral neuropathy: A systematic review and meta-analysis. J Diabetes Investig. 2022 Jan;13(1):134-47. doi: 10.1111/jdi.13643.

10. Pop-Busui R et al. Diabetic neuropathy: A position statement by the American Diabetes Association. Diabetes Care. 2017;40(1):136-54. doi: 10.2337/dc16-2042.

11. Chung YC et al. Calcimimetic restores diabetic peripheral neuropathy by ameliorating apoptosis and improving autophagy. Cell Death Dis. 2018 Nov;9(12):1163. doi: 10.1038/s41419-018-1192-7.

12. Li J et al. Therapeutic effects of moxibustion simultaneously targeting Nrf2 and NF-kB in diabetic peripheral neuropathy. Appl Biochem Biotechnol. 2019 Dec;189(4):1167-82. doi: 10.1007/s12010-019-03052-8.

13. Tan Y et al. Moxibustion for the treatment of diabetic peripheral neuropathy: A systematic review and meta-analysis following PRISMA guidelines. Medicine (Baltimore). 2020 Sep 26;99(39):e22286. doi: 10.1097/MD.0000000000022286.

14. Xie J et al. Protective effect of quercetin on streptozotocin-induced diabetic peripheral neuropathy rats through modulating gut microbiota and reactive oxygen species level. Biomed Pharmacother. 2020 Jul;127:110147. doi: 10.1016/j.biopha.2020.110147.

15. Zhao B et al. Quercetin reduces inflammation in a rat model of diabetic peripheral neuropathy by regulating the TLR4/MyD88/NF-kappa B signalling pathway. Eur J Pharmacol. 2021 Dec;912:174607. doi: 10.1016/j.ejphar.2021.174607.

16. McWhinney SR et al. Platinum neurotoxicity pharmacogenetics. Mol Cancer Ther. 2009;8(1):10-6. doi: 10.1158/1535-7163.MCT-08-0840.

17. Addington J, Freimer M. Chemotherapy-induced peripheral neuropathy: An update on the current understanding. F1000Res. 2016 Jun 22;5:F1000 Faculty Rev-1466. doi: 10.12688/f1000research.8053.1.

18. Lustberg M, Loprinzi C, eds. “Diagnosis, Management and Emerging Strategies for Chemotherapy-Induced Neuropathy: A MASCC Book.” Springer International Publishing; 2021.

19. Cornblath DR et al. Total neuropathy score: Validation and reliability study. Neurology. 1999 Nov;53(8):1660-4. doi: 10.1212/wnl.53.8.1660.

20. Aldossary SA. Review on pharmacology of cisplatin: Clinical use, toxicity and mechanism of resistance of cisplatin. Biomedical and Pharmacology Journal. 2019;12(1):7-15. http://dx.doi.org/10.13005/bpj/1608.

21. Calls A et al. Cisplatin-induced peripheral neuropathy is associated with neuronal senescence-like response. Neuro Oncol. 2021 Jan;23(1):88-99. doi: 10.1093/neuonc/noaa151.

22. Acklin S et al. Depletion of senescent-like neuronal cells alleviates cisplatin-induced peripheral neuropathy in mice. Sci Rep. 2020 Aug;10(1):14170. doi: 10.1038/s41598-020-71042-6.

23. Watanabe S et al. Impact of senescence‐associated secretory phenotype and its potential as a therapeutic target for senescence‐associated diseases. Cancer Sci. 2017 Apr;108(4):563-9. doi: 10.1111/cas.13184.

24. Harrison DE et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009 Jul 16;460(7253):392-5. doi: 10.1038/nature08221.

25. Wakita M et al. A BET family protein degrader provokes senolysis by targeting NHEJ and autophagy in senescent cells. Nat Commun. 2020;11(1):1935. doi: 10.1038/s41467-020-15719-6.

26. Prattichizzo F et al. Anti-TNF-alpha treatment modulates SASP and SASP-related microRNAs in endothelial cells and in circulating angiogenic cells. Oncotarget. 2016 Mar 15;7(11):11945-58. doi: 10.18632/oncotarget.7858.

27. Kang L et al. Oxaliplatin-induced peripheral neuropathy: Clinical features, mechanisms, prevention and treatment. J Neurol. 2021 Sep;268(9):3269-82. doi: 10.1007/s00415-020-09942-w.

28. Yang Y et al. Targeting strategies for oxaliplatin-induced peripheral neuropathy: Clinical syndrome, molecular basis, and drug development. J Exp Clin Cancer Res. 2021 Oct 22;40(1):331. doi: 10.1186/s13046-021-02141-z.

29. Rokhsareh S et al. Evaluating the effects of duloxetine on prophylaxis of oxaliplatin-induced peripheral neuropathy in patients with gastrointestinal cancer: A randomized double-blind placebo controlled clinical trial. J Oncol Pharm Pract. 2021 Nov 5;10781552211052646. doi: 10.1177/10781552211052646.

30. Farshchian N et al. Comparative study of the effects of venlafaxine and duloxetine on chemotherapy-induced peripheral neuropathy. Cancer Chemother Pharmacol. 2018 Nov;82(5):787-93. doi: 10.1007/s00280-018-3664-y.

31. Pereira AF et al. Amifostine protects from the peripheral sensory neuropathy induced by oxaliplatin in mice. Braz J Med Biol Res. 2020 Sep 18;53(11):e10263. doi: 10.1590/1414-431X202010263.

32. Glimelius B et al. Persistent prevention of oxaliplatin-induced peripheral neuropathy using calmangafodipir (PledOx®): A placebo-controlled randomised phase II study (PLIANT). Acta Oncol. 2018 Mar;57(3):393-402. doi: 10.1080/0284186X.2017.1398836.

33. Poupon L et al. Targeting the TREK-1 potassium channel via riluzole to eliminate the neuropathic and depressive-like effects of oxaliplatin. Neuropharmacology. 2018 Sep 15;140:43-61. doi: 10.1016/j.neuropharm.2018.07.026.

34. Mora J et al. Next generation ligand binding assays – Review of emerging technologies’ capabilities to enhance throughput and multiplexing. AAPS J. 2014 Nov;16(6):1175-84. doi: 10.1208/s12248-014-9660-1.

35. Starobova H, Vetter I. Pathophysiology of chemotherapy-induced peripheral neuropathy. Front Mol Neurosci. 2017 May 31;10:174. doi: 10.3389/fnmol.2017.00174.

36. Starobova H et al. Vincristine-induced peripheral neuropathy is driven by canonical NLRP3 activation and IL-1-beta release. J Exp Med. 2021;218(5):e20201452. doi: 10.1084/jem.20201452.

37. Aydin Köker S et al. Effect of pyridoxine plus pyridostigmine treatment on vincristine-induced peripheral neuropathy in pediatric patients with acute lymphoblastic leukemia: A single-center experience. Neurol Sci. 2021 Sep;42(9):3681-6. doi: 10.1007/s10072-020-04970-w.

38. Bourque PR et al. Autoimmune peripheral neuropathies. Clin Chim Acta. 2015 Sep 20;449:37-42. doi: 10.1016/j.cca.2015.02.039.

39. Paparounas K. Anti-GQ1b ganglioside antibody in peripheral nervous system disorders: Pathophysiologic role and clinical relevance. Arch Neurol. 2004 Jul;61(7):1013-6. doi: 10.1001/archneur.61.7.1013.

40. Dalakas MC. Autoimmune peripheral neuropathies, in Rich RR et al., eds., “Clinical Immunology.” 5th ed, (Amsterdam: Elsevier, 2019, pp. 903-915.e1). doi: 10.1016/B978-0-7020-6896-6.00067-3

41. Leonhard SE et al. Diagnosis and management of Guillain-Barré syndrome in ten steps. Nat Rev Neurol. 2019;15(11):671-83. doi: 10.1038/s41582-019-0250-9.

42. Razali SNO et al. Serial peripheral nerve ultrasound in Guillain–Barré syndrome. Clin Neurophysiol. 2016 Nov;127(2):1652-6. doi: 10.1016/j.clinph.2015.06.030.

43. Gallardo E et al. Spinal nerve involvement in early Guillain-Barré syndrome: A clinico-electrophysiological, ultrasonographic and pathological study. Clin Neurophysiol. 2015 Apr;126(4):810-9. doi: 10.1016/j.clinph.2014.06.051.

44. Islam Z et al. Inhibition of C1q, initiator of the classical complement cascade, by ANX005 for the treatment of Guillain–Barré syndrome: Results from a phase 1b study (763). Neurology. 2020 Apr;94(15 Suppl):763.

45. Hughes R et al.; FORCIDP Trial Investigators. Oral fingolimod for chronic inflammatory demyelinating polyradiculoneuropathy (FORCIDP Trial): A double-blind, multicentre, randomised controlled trial. Lancet Neurol. 2018 Aug;17(8):689-98. doi: 10.1016/S1474-4422(18)30202-3.

46. Lansita JA et al. Nonclinical development of ANX005: A humanized anti-C1q antibody for treatment of autoimmune and neurodegenerative diseases. Int J Toxicol. 2017 Nov/Dec;36(6):449-62. doi: 10.1177/1091581817740873.

47. Annexon Inc. A randomized, double-blind, placebo-controlled phase 2/3 study to evaluate the efficacy, safety, pharmacokinetics, and pharmacodynamics of ANX005 in subjects with Guillain–Barré syndrome. ClinicalTrials.gov Identifier: NCT04701164. Updated Jan 8, 2021. Accessed Feb 23, 2022. https://clinicaltrials.gov/ct2/show/NCT04701164.

48. Halstead SK et al. Eculizumab prevents anti-ganglioside antibody-mediated neuropathy in a murine model. Brain. 2008 May;131(Pt 5):1197-1208. doi: 10.1093/brain/awm316.

49. Misawa S et al. Safety and efficacy of eculizumab in Guillain-Barré syndrome: A multicentre, double-blind, randomised phase 2 trial. Lancet Neurol. 2018 Jun;17(6):519-29. doi: 10.1016/S1474-4422(18)30114-5.

50. Alexion Pharmaceuticals. A phase 3, prospective, multicenter, double blind, randomized, placebo-controlled study to evaluate the efficacy and safety of eculizumab in patients with Guillain–Barré syndrome (GBS). ClinicalTrials.gov Identifier: NCT04752566. Updated Feb 18, 2022. Accessed Feb 23, 2022. https://clinicaltrials.gov/ct2/show/NCT04752566.

51. Tzachanis D et al. Successful treatment of refractory Guillain–Barré syndrome with alemtuzumab in a patient with chronic lymphocytic leukemia. Acta Haematol. 2014 Aug;132(2):240-3. doi: 10.1159/000358292.

52. Satkowiak K, Smith AG. Guillain-Barré syndrome, in Roos KL, ed. “Emergency Neurology.” (Springer, Cham, 2021, pp. 225-50). Accessed Feb 23, 2022. https://doi.org/10.1007/978-3-030-75778-6_12.

53. Gogia B et al. Chronic inflammatory demyelinating polyradiculoneuropathy, in “StatPearls [Internet].” (Treasure Island (Fla.): StatPearls Publishing; 2022 Jan). Updated Nov 22, 2021. Accessed Feb 23, 2022. www.ncbi.nlm.nih.gov/books/NBK563249.

54. Allen JA et al. Challenges in the diagnosis of chronic inflammatory demyelinating polyneuropathy. Brain Behav. 2018 Feb;8(3):e00932. doi: 10.1002/brb3.932.

55. Stino AM et al. Chronic inflammatory demyelinating polyradiculoneuropathy-diagnostic pitfalls and treatment approach. Muscle Nerve. 2021 Feb;63(2):157-69. doi: 10.1002/mus.27046.

56. Ginsberg MR et al. Using and interpreting electrodiagnostic tests. Cleve Clin J Med. 2020 Nov 2;87(11):671-82. doi: 10.3949/ccjm.87a.19154.

57. Capodivento G et al. CSF sphingomyelin: A new biomarker of demyelination in the diagnosis and management of CIDP and GBS. J Neurol Neurosurg Psychiatry. 2021;92(3):303-10. doi: 10.1136/jnnp-2020-324445.

58. Shimizu S et al. Efficacy and safety of rituximab in refractory CIDP with or without IgG4 autoantibodies (RECIPE): Protocol for a double-blind, randomized, placebo-controlled clinical trial. JMIR Res Protoc. 2020 Jan 4;9(4):e17117. doi: 10.2196/17117.

59. Plasma Exchange/Sandoglobulin Guillain-Barré Syndrome Trial Group. Randomised trial of plasma exchange, intravenous immunoglobulin, and combined treatments in Guillain-Barré syndrome. Lancet. 1997;349(9047):225-30.

60. Zuercher AW et al. Next-generation Fc receptor–targeting biologics for autoimmune diseases. Autoimmun Rev. 2019 Oct;18(10):102366. doi: 10.1016/j.autrev.2019.102366.

61. Sesarman A et al. The neonatal Fc receptor as therapeutic target in IgG-mediated autoimmune diseases. Cell Mol Life Sci. 2010 Aug;67(15):2533-50. doi: 10.1007/s00018-010-0318-6.

62. Ulrichts P et al. Neonatal Fc receptor antagonist efgartigimod safely and sustainably reduces IgGs in humans. J Clin Invest. 2018 Oct;128(10):4372-86. doi: 10.1172/JCI97911.

63. Peripheral neuropathy [symptoms and causes]. Mayo Clinic [Internet]. Accessed Feb 23, 2022. http://www.mayoclinic.org/diseases-conditions/peripheral-neuropathy/symptoms-causes/syc-20352061.

64. Maharaj SS, Yakasai AM. Does a rehabilitation program of aerobic and progressive resisted exercises influence HIV-induced distal neuropathic pain? Am J Phys Med Rehabil. 2018 May;97(5):364-9. doi: 10.1097/PHM.0000000000000866.

Pages

Next Article: