Archive \ Volume.12 2021 Issue 3

An Overview on the Role of Xanthine Oxidase Inhibitors in Gout Management

, , , , , , , , ,

Abstract

Gout is a kind of inflammatory arthropathy that affects a large number of people. Gout and hyperuricemia are becoming more common in developed countries. Xanthine Oxidase (XO) appears to play a key part in excessive uric acid (UA) production. XO inhibitors medications, which reduce serum UA levels by competitive inhibition of XO, are now the medications of choice for the long-term treatment of hyperuricemia. To discuss the role of XO inhibitors in treating gout disease and provide a review of the different available XO inhibitors medications. For articles selection, PubMed database was utilized, and the following keys were used in the Mesh ((“Xanthine Oxidase Inhibitors”[Mesh]) AND (“Gout” [Mesh]) OR (“Hyperuricemia”[Mesh])). The cornerstone of gout treatment is urea-lowering therapy, which aims to control acute flares, avoid recurring episodes, and prevent or reverse consequences. XO inhibitors such as allopurinol and febuxostat lower urate synthesis and can achieve control of the disease. Allopurinol has been the cornerstone of gout and hyperuricemia clinical therapy, and despite significant tolerability concerns and allegedly low patient compliance, it continues to be the current standard of care. Febuxostat is a safe and effective alternative to allopurinol for people who are allergic to it. However, clinical acceptance of febuxostat has been low, but updated care recommendations recommend febuxostat as an alternative to allopurinol.



How to cite:
Vancouver
Alghamdi AA, Althumali JS, Almalki MMM, Almasoudi AS, Almuntashiri AH, Almuntashiri AH, et al. An Overview on the Role of Xanthine Oxidase Inhibitors in Gout Management. Arch Pharm Pract. 2021;12(3):94-9. https://doi.org/10.51847/RkCPaycprc
APA
Alghamdi, A. A., Althumali, J. S., Almalki, M. M. M., Almasoudi, A. S., Almuntashiri, A. H., Almuntashiri, A. H., Mohammed, A. I., Alkinani, A. A., Almahdawi, M. S., & Mahzari, M. A. H. (2021). An Overview on the Role of Xanthine Oxidase Inhibitors in Gout Management. Archives of Pharmacy Practice, 12(3), 94-99. https://doi.org/10.51847/RkCPaycprc

Download Citation
References

1.        Finch A, Kubler P. The management of gout. Aust Prescr. 2016;39(4):119-22. doi:10.18773/austprescr.2016.047

2.        Graf SW, Whittle SL, Wechalekar MD, Moi JH, Barrett C, Hill CL, et al. Australian and New Zealand recommendations for the diagnosis and management of gout: integrating systematic literature review and expert opinion in the 3e Initiative. Int J Rheum Dis. 2015;18(3):341-51. doi:10.1111/1756-185x.12557

3.        Robinson PC, Horsburgh S. Gout: joints and beyond, epidemiology, clinical features, treatment and co-morbidities. Maturitas. 2014;78(4):245-51. doi:10.1016/j.maturitas.2014.05.001

4.        Zhu Y, Pandya BJ, Choi HK. Comorbidities of gout and hyperuricemia in the US general population: NHANES 2007-2008. Am J Med. 2012;125(7):679-87. doi:10.1016/j.amjmed.2011.09.033

5.        Robinson PC, Taylor WJ, Merriman TR. Systematic review of the prevalence of gout and hyperuricaemia in Australia. Int Med J. 2012;42(9):997-1007. doi:10.1111/j.1445-5994.2012.02794.x

6.        Smith EU, Diaz-Torne C, Perez-Ruiz F, March LM. Epidemiology of gout: an update. Best Pract Res Clin Rheumatol. 2010;24(6):811-27. doi:10.1016/j.berh.2010.10.004

7.        Alghamdi AA, Mutlaqah MA, Amal Mohammed H, Labani LM, Alahmari AF, Abdulrahman R, et al. Gout management in primary care approach; Literature Review. Int J Pharm Res Allied Sci. 2021;10(1):19-23.

8.        Bove M, Cicero AF, Veronesi M, Borghi C. An evidence-based review on urate-lowering treatments: implications for optimal treatment of chronic hyperuricemia. Vasc Health Risk Manag. 2017;13:23-8. doi:10.2147/vhrm.s115080

9.        Viggiano D, Gigliotti G, Vallone G, Giammarino A, Nigro M, Capasso G. Urate-lowering agents in asymptomatic hyperuricemia: role of urine sediment analysis and musculoskeletal ultrasound. Kidney Blood Press Res. 2018;43(2):606-15. doi:10.1159/000489145

10.      Watanabe S, Kang DH, Feng L, Nakagawa T, Kanellis J, Lan H, et al. Uric acid, hominoid evolution, and the pathogenesis of salt-sensitivity. Hypertension. 2002;40(3):355-60. doi:10.1161/01.hyp.0000028589.66335.aa

11.      Viggiano A, Viggiano D, Viggiano A, Luca BD. Quantitative histochemical assay for superoxide dismutase in rat brain. J Histochem Cytochem. 2003;51(7):865-71. doi:10.1177/002215540305100702

12.      Perna AF, Di Nunzio A, Amoresano A, Pane F, Fontanarosa C, Pucci P, et al. Divergent behavior of hydrogen sulfide pools and of the sulfur metabolite lanthionine, a novel uremic toxin, in dialysis patients. Biochimie. 2016;126:97-107. doi:10.1016/j.biochi.2016.04.018

13.      Ragab G, Elshahaly M, Bardin T. Gout: An old disease in new perspective–A review. J Adv Res. 2017;8(5):495-511. doi:10.1016/j.jare.2017.04.008

14.      Dalbeth N, Merriman TR, Stamp LK. Gout. Lancet. 2016;388(10055):2039-52. doi:10.1016/s0140-6736(16)00346-9

15.      Mandal AK, Mount DB. The molecular physiology of uric acid homeostasis. Annu Rev Physiol. 2015;77(1):323-45. doi:10.1146/annurev-physiol-021113-170343

16.      Kamei K, Konta T, Hirayama A, Suzuki K, Ichikawa K, Fujimoto S, et al. A slight increase within the normal range of serum uric acid and the decline in renal function: associations in a community-based population. Nephrol Dial Transplant. 2014;29(12):2286-92. doi:10.1093/ndt/gfu256

17.      Torres RJ, Puig JG. Hypoxanthine-guanine phosophoribosyltransferase (HPRT) deficiency: Lesch-Nyhan syndrome. Orphanet J Rare Dis. 2007;2(1):1-0. doi:10.1186/1750-1172-2-48

18.      Kanbara A, Seyama I. Effect of urine pH on uric acid excretion by manipulating food materials. Nucleosides Nucleotides Nucleic Acids. 2011;30(12):1066-71. doi:10.1080/15257770.2011.596498

19.      Dessein PH, Shipton EA, Stanwix AE, Joffe BI, Ramokgadi J. Beneficial effects of weight loss associated with moderate calorie/carbohydrate restriction, and increased proportional intake of protein and unsaturated fat on serum urate and lipoprotein levels in gout: a pilot study. Ann Rheum Dis. 2000;59(7):539-43. doi:10.1136/ard.59.7.539

20.      Ichida K, Matsuo H, Takada T, Nakayama A, Murakami K, Shimizu T, et al. Decreased extra-renal urate excretion is a common cause of hyperuricemia. Nat Commun. 2012;3(1):1-7. doi:10.1038/ncomms1756

21.      Bobulescu IA, Moe OW. Renal transport of uric acid: evolving concepts and uncertainties. Adv Chronic Kidney Dis. 2012;19(6):358-71. doi:10.1053/j.ackd.2012.07.009

22.      Han J, Liu Y, Rao F, Nievergelt CM, O'Connor DT, Wang X, et al. Common genetic variants of the human uromodulin gene regulate transcription and predict plasma uric acid levels. Kidney Int. 2013;83(4):733-40. doi:10.1038/ki.2012.449

23.      Perez-Ruiz F, Dalbeth N, Bardin T. A review of uric acid, crystal deposition disease, and gout. Adv Ther. 2015;32(1):31-41. doi:10.1007/s12325-014-0175-z

24.      Robinson PC, Dalbeth N. Advances in pharmacotherapy for the treatment of gout. Expert Opin Pharmacother. 2015;16(4):533-46. doi:10.1517/14656566.2015.997213

25.      Strilchuk L, Fogacci F, Cicero AF. Safety and tolerability of available urate-lowering drugs: a critical review. Expert Opin Drug Saf. 2019;18(4):261-71. doi:10.1080/14740338.2019.1594771

26.      Pittman JR, Bross MH. Diagnosis and management of gout. Am Fam Physician. 1999;59(7):1799-806.

27.      Khanna D, Fitzgerald JD, Khanna PP, Bae S, Singh MK, Neogi T, et al. 2012 American College of Rheumatology guidelines for management of gout. Part 1: systematic nonpharmacologic and pharmacologic therapeutic approaches to hyperuricemia. Arthritis Care Res. 2012;64(10):1431-46. doi:10.1002/acr.21772

28.      Rees F, Hui M, Doherty M. Optimizing current treatment of gout. Nat Rev Rheumatol. 2014;10(5):271-83. doi:10.1038/nrrheum.2014.32

29.      Sundy JS, Ganson NJ, Kelly SJ, Scarlett EL, Rehrig CD, Huang W, et al. Pharmacokinetics and pharmacodynamics of intravenous PEGylated recombinant mammalian urate oxidase in patients with refractory gout. Arthritis Rheum. 2007;56(3):1021-8. doi:10.1002/art.22403

30.      Day RO, Graham GG, Hicks M, McLachlan AJ, Stocker SL, Williams KM. Clinical pharmacokinetics and pharmacodynamics of allopurinol and oxypurinol. Clin Pharmacokinet. 2007;46(8):623-44. doi:10.2165/00003088-200746080-00001

31.      Stamp LK, Taylor WJ, Jones PB, Dockerty JL, Drake J, Frampton C, et al. Starting dose is a risk factor for allopurinol hypersensitivity syndrome: a proposed safe starting dose of allopurinol. Arthritis Rheum. 2012;64(8):2529-36. doi:10.1002/art.34488

32.      Ghislain PD, Roujeau JC. Treatment of severe drug reactions: Stevens-Johnson syndrome, toxic epidermal necrolysis and hypersensitivity syndrome. Dermatol Online J. 2002;8(1). doi:10.5070/d397d8t291

33.      Stamp LK, Barclay ML. How to prevent allopurinol hypersensitivity reactions?. Rheumatology. 2018;57(suppl_1):i35-41. doi:10.1093/rheumatology/kex422

34.      Ramasamy SN, Korb-Wells CS, Kannangara DR, Smith MW, Wang N, Roberts DM, et al. Allopurinol hypersensitivity: a systematic review of all published cases, 1950–2012. Drug Saf. 2013;36(10):953-80. doi:10.1007/s40264-013-0084-0

35.      Benn CL, Dua P, Gurrell R, Loudon P, Pike A, Storer RI, et al. Physiology of hyperuricemia and urate-lowering treatments. Front Med. 2018;5:160. doi:10.3389/fmed.2018.00160

36.      Caulfield MJ, Munroe PB, O'Neill D, Witkowska K, Charchar FJ, Doblado M, et al. SLC2A9 is a high-capacity urate transporter in humans. PLoS Med. 2008;5(10):e197. doi:10.1371/journal.pmed.0050197

37.      Huang CH, Chu MP, Chen WW. Examining the use of allopurinol: perspectives from recent drug injury relief applications. J Formos Med Assoc. 2019;118(1):371-7. doi:10.1016/j.jfma.2018.06.006

38.      Pascual E, Sivera F, Yasothan U, Kirkpatrick P. Febuxostat. Nat Rev Drug Discov. 2009;8(3):191-3. doi:10.1038/nrd2831

39.      Borghi C, Cicero AF. Serum Uric Acid and Cardiometabolic Disease. Hypertension. 2017;69(6):1011-3. doi:10.1161/hypertensionaha.117.09081

40.      Schumacher Jr HR, Becker MA, Wortmann RL, MacDonald PA, Hunt B, Streit J, et al. Effects of febuxostat versus allopurinol and placebo in reducing serum urate in subjects with hyperuricemia and gout: a 28‐week, phase III, randomized, double‐blind, parallel‐group trial. Arthritis Care Res. 2008;59(11):1540-8. doi:10.1002/art.24209

41.      White WB, Saag KG, Becker MA, Borer JS, Gorelick PB, Whelton A, et al. Cardiovascular safety of febuxostat or allopurinol in patients with gout. N Eng J Med. 2018;378(13):1200-10. doi:10.1056/nejmoa1710895

42.      Katsiki N, Borghi C. The future of febuxostat after the Cardiovascular Safety of Febuxostat and Allopurinol in Patients with Gout and Cardiovascular Morbidities (CARES) trial: who CARES?. Expert Opin Pharmacother. 2018;19(17):1853-6. doi:10.1080/14656566.2018.1532503

43.      Jordan A, Gresser U. Side effects and interactions of the xanthine oxidase inhibitor febuxostat. Pharmaceuticals. 2018;11(2):51. doi:10.3390/ph11020051

44.      Hosoya T, Ogawa Y, Hashimoto H, Ohashi T, Sakamoto R. Comparison of topiroxostat and allopurinol in Japanese hyperuricemic patients with or without gout: A phase 3, multicentre, randomized, double‐blind, double‐dummy, active‐controlled, parallel‐group study. J Clin Pharm Ther. 2016;41(3):290-7. doi:10.1111/jcpt.12391

45.      Horino T, Hatakeyama Y, Ichii O, Matsumoto T, Shimamura Y, Inoue K, et al. Effects of topiroxostat in hyperuricemic patients with chronic kidney disease. Clin Exp Nephrol. 2018;22(2):337-45. doi:10.1007/s10157-017-1452-3

46.      Sato T, Ashizawa N, Matsumoto K, Iwanaga T, Nakamura H, Inoue T, et al. Discovery of 3-(3-cyano-4-pyridyl)-5-(4-pyridyl)-1, 2, 4-triazole, FYX-051-a xanthine oxidoreductase inhibitor for the treatment of hyperuricemia. Bioorg Med Chem Lett. 2009;19(21):6225-9. doi:10.1016/j.bmcl.2009.08.091

47.      Hosoya T, Sasaki T, Ohashi T. Clinical efficacy and safety of topiroxostat in Japanese hyperuricemic patients with or without gout: a randomized, double-blinded, controlled phase 2b study. Clin Rheumtol. 2017;36(3):649-56. doi:10.1007/s10067-016-3474-8

48.      Greig D, Alcaino H, Castro PF, Garcia L, Verdejo HE, Navarro M, et al. Xanthine-oxidase inhibitors and statins in chronic heart failure: effects on vascular and functional parameters. J Heart Lung Transplant. 2011;30(4):408-13. doi:10.1016/j.healun.2010.10.003

49.      Mizukoshi T, Kato S, Ando M, Sobajima H, Ohashi N, Naruse T, et al. Renoprotective effects of topiroxostat for hyperuricaemic patients with overt diabetic nephropathy study (ETUDE study): a prospective, randomized, multicentre clinical trial. Nephrology. 2018;23(11):1023-30. doi:10.1111/nep.13177

Sezai A, Obata K, Abe K, Kanno S, Sekino H. Cross-over trial of febuxostat and topiroxostat for hyperuricemia with cardiovascular disease (TROFEO trial). Circ J. 2017;81(11):1707-12. doi:10.1253/circj.cj-17-0438


Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.