Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Primer
  • Published:

Gout

Subjects

Abstract

Gout is a chronic disease caused by monosodium urate (MSU) crystal deposition. Gout typically presents as an acute, self-limiting inflammatory monoarthritis that affects the joints of the lower limb. Elevated serum urate level (hyperuricaemia) is the major risk factor for MSU crystal deposition and development of gout. Although traditionally considered a disorder of purine metabolism, altered urate transport, both in the gut and the kidneys, has a key role in the pathogenesis of hyperuricaemia. Anti-inflammatory agents, such corticosteroids, NSAIDs and colchicine, are widely used for the treatment of gout flare; recognition of the importance of NLRP3 inflammasome activation and bioactive IL-1β release in initiation of the gout flare has led to the development of anti-IL-1β biological therapy for gout flares. Sustained reduction in serum urate levels using urate-lowering therapy is vital in the long-term management of gout, which aims to dissolve MSU crystals, suppress gout flares and resolve tophi. Allopurinol is the first-line urate-lowering therapy and should be started at a low dose, with gradual dose escalation. Low-dose anti-inflammatory therapies can reduce gout flares during initiation of urate-lowering therapy. Models of care, such as nurse-led strategies that focus on patient engagement and education, substantially improve clinical outcomes and now represent best practice for gout management.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Disease progression in gout.
Fig. 2: Urate transporters in the kidneys and intestine in humans.
Fig. 3: Classification of the causes of hyperuricaemia.
Fig. 4: Mechanisms of initiation and resolution of the gout flare.
Fig. 5: Identification of MSU crystals using polarizing light microscopy.
Fig. 6: Typical clinical and imaging features of gout.
Fig. 7: Ultrasonography-based features of gout.
Fig. 8: Mechanism of action of urate-lowering drugs currently used for gout treatment.

Similar content being viewed by others

References

  1. Chen-Xu, M., Yokose, C., Rai, S. K., Pillinger, M. H. & Choi, H. K. Contemporary prevalence of gout and hyperuricemia in the United States and decadal trends: the National Health and Nutrition Examination Survey 2007–2016. Arthritis Rheumatol. 71, 991–999 (2019). This article describes the contemporary epidemiology of gout and hyperuricaemia in the USA.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Faires, J. S. & McCarty, D. J. Acute arthritis in man and dog after intrasynovial injection of sodium urate crystals. Lancet 280, 682–685 (1962). This paper proved that monosodium urate crystals cause the gout flare.

    Article  Google Scholar 

  3. Loeb, J. N. The influence of temperature on the solubility of monosodium urate. Arthritis Rheum. 15, 189–192 (1972).

    Article  CAS  PubMed  Google Scholar 

  4. Taylor, W. J. et al. Study for updated gout classification criteria: identification of features to classify gout. Arthritis Care Res. (Hoboken) 67, 1304–1315 (2015). This study describes the major clinical features of gout in a large multicentre study, using monosodium urate crystal identification as the gold standard.

    Article  Google Scholar 

  5. Gutman, A. B. The past four decades of progress in the knowledge of gout, with an assessment of the present status. Arthritis Rheum. 16, 431–445 (1973).

    Article  CAS  PubMed  Google Scholar 

  6. Neogi, T. et al. 2015 gout classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Ann. Rheum. Dis. 74, 1789–1798 (2015). This paper describes the 2015 ACR/EULAR gout classification criteria.

    Article  CAS  PubMed  Google Scholar 

  7. Bursill, D. et al. Gout, Hyperuricemia, and Crystal-Associated Disease Network consensus statement regarding labels and definitions for disease elements in gout. Arthritis Care Res. (Hoboken) 71, 427–434 (2019).

    Article  Google Scholar 

  8. Kuo, C. F., Grainge, M. J., Zhang, W. & Doherty, M. Global epidemiology of gout: prevalence, incidence and risk factors. Nat. Rev. Rheumatol. 11, 649–662 (2015).

    Article  PubMed  Google Scholar 

  9. Zhu, Y., Pandya, B. J. & Choi, H. K. Prevalence of gout and hyperuricemia in the US general population: the National Health and Nutrition Examination Survey 2007–2008. Arthritis Rheum. 63, 3136–3141 (2011).

    Article  PubMed  Google Scholar 

  10. Lawrence, R. C. et al. Estimates of the prevalence of selected arthritic and musculoskeletal diseases in the United States. J. Rheumatol. 16, 427–441 (1989).

    CAS  PubMed  Google Scholar 

  11. Choi, H. K. & Curhan, G. Soft drinks, fructose consumption, and the risk of gout in men: prospective cohort study. BMJ 336, 309–312 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Choi, H. K., Willett, W. & Curhan, G. Fructose-rich beverages and risk of gout in women. JAMA 304, 2270–2278 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Currie, W. J. Prevalence and incidence of the diagnosis of gout in Great Britain. Ann. Rheum. Dis. 38, 101–106 (1979).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Harris, C. M., Lloyd, D. C. & Lewis, J. The prevalence and prophylaxis of gout in England. J. Clin. Epidemiol. 48, 1153–1158 (1995).

    Article  CAS  PubMed  Google Scholar 

  15. Kuo, C. F., Grainge, M. J., Mallen, C., Zhang, W. & Doherty, M. Rising burden of gout in the UK but continuing suboptimal management: a nationwide population study. Ann. Rheum. Dis. 74, 661–667 (2015).

    Article  PubMed  Google Scholar 

  16. Zhang, Y. et al. Alcohol consumption as a trigger of recurrent gout attacks. Am. J. Med. 119, 800.e11–800.e16 (2006).

    Article  CAS  Google Scholar 

  17. Kuo, C. F. et al. Epidemiology and management of gout in Taiwan: a nationwide population study. Arthritis Res. Ther. 17, 13 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Roddy, E. & Doherty, M. Epidemiology of gout. Arthritis Res. Ther. 12, 223 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Arromdee, E., Michet, C. J., Crowson, C. S., O’Fallon, W. M. & Gabriel, S. E. Epidemiology of gout: is the incidence rising? J. Rheumatol. 29, 2403–2406 (2002).

    PubMed  Google Scholar 

  20. Nakayama, A. et al. GWAS of clinically defined gout and subtypes identifies multiple susceptibility loci that include urate transporter genes. Ann. Rheum. Dis. 76, 869–877 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Lim, S. Y. et al. Trends in gout and rheumatoid arthritis hospitalizations in the United States, 1993-2011. JAMA 315, 2345–2347 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Robinson, P. C., Merriman, T. R., Herbison, P. & Highton, J. Hospital admissions associated with gout and their comorbidities in New Zealand and England 1999–2009. Rheumatology 52, 118–126 (2013).

    Article  PubMed  Google Scholar 

  23. Dehlin, M. & Jacobsson, L. T. H. Trends in gout hospitalization in Sweden. J. Rheumatol. 45, 145–146 (2018).

    Article  PubMed  Google Scholar 

  24. Edwards, N. L. Quality of care in patients with gout: why is management suboptimal and what can be done about it? Curr. Rheumatol. Rep. 13, 154–159 (2011).

    Article  PubMed  Google Scholar 

  25. Sarawate, C. A. et al. Gout medication treatment patterns and adherence to standards of care from a managed care perspective. Mayo Clin. Proc. 81, 925–934 (2006).

    Article  PubMed  Google Scholar 

  26. Shiozawa, A., Szabo, S. M., Bolzani, A., Cheung, A. & Choi, H. K. Serum uric acid and the risk of incident and recurrent gout: a systematic review. J. Rheumatol. 44, 388–396 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Rothenbacher, D., Primatesta, P., Ferreira, A., Cea-Soriano, L. & Rodriguez, L. A. Frequency and risk factors of gout flares in a large population-based cohort of incident gout. Rheumatology 50, 973–981 (2011).

    Article  PubMed  Google Scholar 

  28. Zhang, Y. et al. Purine-rich foods intake and recurrent gout attacks. Ann. Rheum. Dis. 71, 1448–1453 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Dubreuil, M. et al. Increased risk of recurrent gout attacks with hospitalization. Am. J. Med. 126, 1138–1141.e1 (2013).

    Article  PubMed  Google Scholar 

  30. Hak, A. E., Curhan, G. C., Grodstein, F. & Choi, H. K. Menopause, postmenopausal hormone use and risk of incident gout. Ann. Rheum. Dis. 69, 1305–1309 (2010).

    Article  PubMed  Google Scholar 

  31. Puig, J. G. et al. Female gout. Clinical spectrum and uric acid metabolism. Arch. Intern. Med. 151, 726–732 (1991).

    Article  CAS  PubMed  Google Scholar 

  32. Dalbeth, N. et al. Relationship between serum urate concentration and clinically evident incident gout: an individual participant data analysis. Ann. Rheum. Dis. 77, 1048–1052 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Campion, E. W., Glynn, R. J. & DeLabry, L. O. Asymptomatic hyperuricemia. Risks and consequences in the Normative Aging Study. Am. J. Med. 82, 421–426 (1987).

    Article  CAS  PubMed  Google Scholar 

  34. Kapetanovic, M. C. et al. The risk of clinically diagnosed gout by serum urate levels: results from 30 years follow-up of the Malmo Preventive Project Cohort in Southern Sweden. Arthritis Res. Ther. 20, 190 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Dalbeth, N. et al. Urate crystal deposition in asymptomatic hyperuricaemia and symptomatic gout: a dual energy CT study. Ann. Rheum. Dis. 74, 908–911 (2015).

    Article  PubMed  Google Scholar 

  36. De Miguel, E. et al. Diagnosis of gout in patients with asymptomatic hyperuricaemia: a pilot ultrasound study. Ann. Rheum. Dis. 71, 157–158 (2012).

    Article  PubMed  Google Scholar 

  37. Dalbeth, N. et al. Cellular characterization of the gouty tophus: a quantitative analysis. Arthritis Rheum. 62, 1549–1556 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Wu, X. W., Lee, C. C., Muzny, D. M. & Caskey, C. T. Urate oxidase: primary structure and evolutionary implications. Proc. Natl Acad. Sci. USA 86, 9412–9416 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ames, B. N., Cathcart, R., Schwiers, E. & Hochstein, P. Uric acid provides an antioxidant defense in humans against oxidant- and radical-caused aging and cancer: a hypothesis. Proc. Natl Acad. Sci. USA 78, 6858–6862 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Shi, Y., Evans, J. E. & Rock, K. L. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature 425, 516–521 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. Watanabe, S. et al. Uric acid, hominoid evolution, and the pathogenesis of salt-sensitivity. Hypertension 40, 355–360 (2002).

    Article  CAS  PubMed  Google Scholar 

  42. Choi, H. K., Liu, S. & Curhan, G. Intake of purine-rich foods, protein, and dairy products and relationship to serum levels of uric acid: the Third National Health and Nutrition Examination Survey. Arthritis Rheum. 52, 283–289 (2005).

    Article  PubMed  Google Scholar 

  43. Choi, H. K., Atkinson, K., Karlson, E. W., Willett, W. & Curhan, G. Purine-rich foods, dairy and protein intake, and the risk of gout in men. N. Engl. J. Med. 350, 1093–1103 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. Stirpe, F. et al. Fructose-induced hyperuricaemia. Lancet 2, 1310–1311 (1970).

    Article  CAS  PubMed  Google Scholar 

  45. Bode, C., Schumacher, H., Goebell, H., Zelder, O. & Pelzel, H. Fructose induced depletion of liver adenine nucleotides in man. Hormone Metab. Res. 3, 289–290 (1971).

    Article  CAS  Google Scholar 

  46. Choi, J. W., Ford, E. S., Gao, X. & Choi, H. K. Sugar-sweetened soft drinks, diet soft drinks, and serum uric acid level: the Third National Health and Nutrition Examination Survey. Arthritis Rheum. 59, 109–116 (2008).

    Article  CAS  PubMed  Google Scholar 

  47. Faller, J. & Fox, I. H. Ethanol-induced hyperuricemia: evidence for increased urate production by activation of adenine nucleotide turnover. N. Engl. J. Med. 307, 1598–1602 (1982).

    Article  CAS  PubMed  Google Scholar 

  48. Puig, J. G. & Fox, I. H. Ethanol-induced activation of adenine nucleotide turnover. Evidence for a role of acetate. J. Clin. Invest. 74, 936–941 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Choi, H. K. & Curhan, G. Coffee, tea, and caffeine consumption and serum uric acid level: the Third National Health and Nutrition Examination Survey. Arthritis Rheum. 57, 816–821 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Gao, X., Curhan, G., Forman, J. P., Ascherio, A. & Choi, H. K. Vitamin C intake and serum uric acid concentration in men. J. Rheumatol. 35, 1853–1858 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Choi, H. K., Soriano, L. C., Zhang, Y. & Rodriguez, L. A. Antihypertensive drugs and risk of incident gout among patients with hypertension: population based case-control study. BMJ 344, d8190 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Zhu, Y., Pandya, B. J. & Choi, H. K. Comorbidities of gout and hyperuricemia in the US general population: NHANES 2007-2008. Am. J. Med. 125, 679–687.e1 (2012).

    Article  PubMed  Google Scholar 

  53. Nakayama, A. et al. Common dysfunctional variants of ABCG2 have stronger impact on hyperuricemia progression than typical environmental risk factors. Sci. Rep. 4, 5227 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Major, T. J., Topless, R. K., Dalbeth, N. & Merriman, T. R. Evaluation of the diet wide contribution to serum urate levels: meta-analysis of population based cohorts. BMJ 363, k3951 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Perez-Ruiz, F., Calabozo, M., Erauskin, G. G., Ruibal, A. & Herrero-Beites, A. M. Renal underexcretion of uric acid is present in patients with apparent high urinary uric acid output. Arthritis Rheum. 47, 610–613 (2002).

    Article  CAS  PubMed  Google Scholar 

  56. Kottgen, A. et al. Genome-wide association analyses identify 18 new loci associated with serum urate concentrations. Nat. Genet. 45, 145–154 (2013). This landmark paper describes a large GWAS to identify loci associated with serum urate levels and gout, emphasizing the importance of urate transporters and glucose metabolism in regulation of serum urate concentrations.

    Article  PubMed  CAS  Google Scholar 

  57. Nakatochi, M. et al. Genome-wide meta-analysis identifies multiple novel loci associated with serum uric acid levels in Japanese individuals. Commun. Biol. 2, 115 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Li, C. et al. Genome-wide association analysis identifies three new risk loci for gout arthritis in Han Chinese. Nat. Commun. 6, 7041 (2015).

    Article  CAS  PubMed  Google Scholar 

  59. Phipps-Green, A. J. et al. Twenty-eight loci that influence serum urate levels: analysis of association with gout. Ann. Rheum. Dis. 75, 124–130 (2016).

    Article  CAS  PubMed  Google Scholar 

  60. Matsuo, H. et al. Genome-wide association study of clinically defined gout identifies multiple risk loci and its association with clinical subtypes. Ann. Rheum. Dis. 75, 652–659 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. Enomoto, A. et al. Molecular identification of a renal urate anion exchanger that regulates blood urate levels. Nature 417, 447–452 (2002). This paper describes the discovery of URAT1, the major target of uricosuric agents.

    Article  CAS  PubMed  Google Scholar 

  62. Shin, H. J. et al. Interactions of urate transporter URAT1 in human kidney with uricosuric drugs. Nephrology 16, 156–162 (2011).

    Article  CAS  PubMed  Google Scholar 

  63. Miner, J. N. et al. Lesinurad, a novel, oral compound for gout, acts to decrease serum uric acid through inhibition of urate transporters in the kidney. Arthritis Res. Ther. 18, 214 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Matsuo, H. et al. Mutations in glucose transporter 9 gene SLC2A9 cause renal hypouricemia. Am. J. Hum. Genet. 83, 744–751 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Dinour, D. et al. Homozygous SLC2A9 mutations cause severe renal hypouricemia. J. Am. Soc. Nephrol. 21, 64–72 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Kawamura, Y. et al. Pathogenic GLUT9 mutations causing renal hypouricemia type 2 (RHUC2). Nucleosides Nucleotides Nucleic Acids 30, 1105–1111 (2011).

    Article  CAS  PubMed  Google Scholar 

  67. Chiba, T. et al. NPT1/SLC17A1 is a renal urate exporter in humans and its common gain-of-function variant decreases the risk of renal underexcretion gout. Arthritis Rheumatol. 67, 281–287 (2015).

    Article  CAS  PubMed  Google Scholar 

  68. Hollis-Moffatt, J. E. et al. The renal urate transporter SLC17A1 locus: confirmation of association with gout. Arthritis Res. Ther. 14, R92 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Woodward, O. M. et al. Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc. Natl Acad. Sci. USA 106, 10338–10342 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Matsuo, H. et al. Common defects of ABCG2, a high-capacity urate exporter, cause gout: a function-based genetic analysis in a Japanese population. Sci. Transl Med. 1, 5ra11 (2009).

    Article  PubMed  CAS  Google Scholar 

  71. Matsuo, H. et al. ABCG2 dysfunction causes hyperuricemia due to both renal urate underexcretion and renal urate overload. Sci. Rep. 4, 3755 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Matsuo, H. et al. Hyperuricemia in acute gastroenteritis is caused by decreased urate excretion via ABCG2. Sci. Rep. 6, 31003 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Matsuo, H. et al. Common dysfunctional variants in ABCG2 are a major cause of early-onset gout. Sci. Rep. 3, 2014 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Phipps-Green, A. J. et al. A strong role for the ABCG2 gene in susceptibility to gout in New Zealand Pacific Island and Caucasian, but not Maori, case and control sample sets. Hum. Mol. Genet. 19, 4813–4819 (2010).

    Article  CAS  PubMed  Google Scholar 

  75. Higashino, T. et al. Multiple common and rare variants of ABCG2 cause gout. RMD Open 3, e000464 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Stiburkova, B. et al. Functional non-synonymous variants of ABCG2 and gout risk. Rheumatology 56, 1982–1992 (2017).

    Article  CAS  PubMed  Google Scholar 

  77. Dehghan, A. et al. Association of three genetic loci with uric acid concentration and risk of gout: a genome-wide association study. Lancet 372, 1953–1961 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Ichida, K. et al. Decreased extra-renal urate excretion is a common cause of hyperuricemia. Nat. Commun. 3, 764 (2012). This paper describes decreased extra-renal urate excretion caused by ABCG2 dysfunction as a new concept in the pathogenesis of hyperuricaemia.

    Article  PubMed  CAS  Google Scholar 

  79. Becker, M. A. in The Metabolic and Molecular Bases of Inherited Disease (eds Scriver, C. R., Childs, B., Kinzler, K. W., & Vogelstein, B.) Ch. 106, 2513–2535 (McGraw-Hill, 2001).

  80. Wortmann, R. L. in Harrison’s Principles of Internal Medicine (eds Fauci, A. S. et al.) Ch. 353, 2444–2449 (McGraw-Hill, 2008).

  81. Mandel, N. S. & Mandel, G. S. Monosodium urate monohydrate, the gout culprit. J. Am. Chem. Soc. 98, 2319–2323 (1976).

    Article  CAS  PubMed  Google Scholar 

  82. Dalbeth, N. et al. Tendon involvement in the feet of patients with gout: a dual-energy CT study. Ann. Rheum. Dis. 72, 1545–1548 (2013).

    Article  PubMed  Google Scholar 

  83. Chhana, A., Lee, G. & Dalbeth, N. Factors influencing the crystallization of monosodium urate: a systematic literature review. BMC Musculoskelet. Disord. 16, 296 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Schumacher, H. R. Pathology of the synovial membrane in gout. Light and electron microscopic studies. Interpretation of crystals in electron micrographs. Arthritis Rheum. 18, 771–782 (1975).

    Article  CAS  PubMed  Google Scholar 

  85. Wood, D. D., Ihrie, E. J., Dinarello, C. A. & Cohen, P. L. Isolation of an interleukin-1-like factor from human joint effusions. Arthritis Rheum. 26, 975–983 (1983).

    Article  CAS  PubMed  Google Scholar 

  86. Di Giovine, F. S., Malawista, S. E., Nuki, G. & Duff, G. W. Interleukin 1 (IL 1) as a mediator of crystal arthritis. Stimulation of T cell and synovial fibroblast mitogenesis by urate crystal-induced IL 1. J. Immunol. 138, 3213–3218 (1987).

    PubMed  Google Scholar 

  87. Martinon, F., Petrilli, V., Mayor, A., Tardivel, A. & Tschopp, J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440, 237–241 (2006). This paper describes the crucial role of activation of the NLRP3 inflammasome and release of mature IL-1β in the initiation of the gout flare.

    Article  CAS  PubMed  Google Scholar 

  88. Solomon, D. H. et al. Relationship of interleukin-1β blockade with incident gout and serum uric acid levels: exploratory analysis of a randomized controlled trial. Ann. Intern. Med. 169, 535–542 (2018).

    Article  PubMed  Google Scholar 

  89. Schlesinger, N. et al. Canakinumab reduces the risk of acute gouty arthritis flares during initiation of allopurinol treatment: results of a double-blind, randomised study. Ann. Rheum. Dis. 70, 1264–1271 (2011).

    Article  CAS  PubMed  Google Scholar 

  90. Janssen, C. A. et al. Anakinra for the treatment of acute gout flares: a randomized, double-blind, placebo-controlled, active-comparator, non-inferiority trial. Rheumatology 58, 1344–1352 (2019).

    Article  Google Scholar 

  91. Schlesinger, N. et al. Canakinumab for acute gouty arthritis in patients with limited treatment options: results from two randomised, multicentre, active-controlled, double-blind trials and their initial extensions. Ann. Rheum. Dis. 71, 1839–1848 (2012).

    Article  CAS  PubMed  Google Scholar 

  92. Martinon, F., Burns, K. & Tschopp, J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol. Cell 10, 417–426 (2002).

    Article  CAS  PubMed  Google Scholar 

  93. Pascual, E., Batlle-Gualda, E., Martinez, A., Rosas, J. & Vela, P. Synovial fluid analysis for diagnosis of intercritical gout. Ann. Intern. Med. 131, 756–759 (1999).

    Article  CAS  PubMed  Google Scholar 

  94. Mangan, M. S. J. et al. Targeting the NLRP3 inflammasome in inflammatory diseases. Nat. Rev. Drug Discov. 17, 588–606 (2018).

    Article  CAS  PubMed  Google Scholar 

  95. Giamarellos-Bourboulis, E. J. et al. Crystals of monosodium urate monohydrate enhance lipopolysaccharide-induced release of interleukin 1 beta by mononuclear cells through a caspase 1-mediated process. Ann. Rheum. Dis. 68, 273–278 (2009).

    Article  CAS  PubMed  Google Scholar 

  96. Joosten, L. A. et al. Engagement of fatty acids with Toll-like receptor 2 drives interleukin-1β production via the ASC/caspase 1 pathway in monosodium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum. 62, 3237–3248 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Crisan, T. O. et al. Soluble uric acid primes TLR-induced proinflammatory cytokine production by human primary cells via inhibition of IL-1Ra. Ann. Rheum. Dis. 75, 755–762 (2016).

    Article  CAS  PubMed  Google Scholar 

  98. Mylona, E. E. et al. Enhanced interleukin-1β production of PBMCs from patients with gout after stimulation with Toll-like receptor-2 ligands and urate crystals. Arthritis Res. Ther. 14, R158 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Crisan, T. O. et al. Uric acid priming in human monocytes is driven by the AKT-PRAS40 autophagy pathway. Proc. Natl Acad. Sci. USA 114, 5485–5490 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Chen, Y. H. et al. Spontaneous resolution of acute gouty arthritis is associated with rapid induction of the anti-inflammatory factors TGFβ1, IL-10 and soluble TNF receptors and the intracellular cytokine negative regulators CIS and SOCS3. Ann. Rheum. Dis. 70, 1655–1663 (2011).

    Article  CAS  PubMed  Google Scholar 

  101. Yagnik, D. R. et al. Macrophage release of transforming growth factor beta1 during resolution of monosodium urate monohydrate crystal-induced inflammation. Arthritis Rheum. 50, 2273–2280 (2004).

    Article  CAS  PubMed  Google Scholar 

  102. Barden, A. E. et al. Specialised pro-resolving mediators of inflammation in inflammatory arthritis. Prostaglandins Leukot. Essent. Fatty Acids 107, 24–29 (2016).

    Article  CAS  PubMed  Google Scholar 

  103. Liu, L. et al. Interleukin 37 limits monosodium urate crystal-induced innate immune responses in human and murine models of gout. Arthritis Res. Ther. 18, 268 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Steiger, S. & Harper, J. L. Neutrophil cannibalism triggers transforming growth factor β1 production and self regulation of neutrophil inflammatory function in monosodium urate monohydrate crystal-induced inflammation in mice. Arthritis Rheum. 65, 815–823 (2013).

    Article  CAS  PubMed  Google Scholar 

  105. Schauer, C. et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat. Med. 20, 511–517 (2014). This article describes the role of aggNETs in resolution of the gout flare and also implicates aggNETs in development of the tophus.

    Article  CAS  PubMed  Google Scholar 

  106. Sokoloff, L. The pathology of gout. Metabolism 6, 230–243 (1957).

    CAS  PubMed  Google Scholar 

  107. Schweyer, S., Hemmerlein, B., Radzun, H. J. & Fayyazi, A. Continuous recruitment, co-expression of tumour necrosis factor-alpha and matrix metalloproteinases, and apoptosis of macrophages in gout tophi. Virchows Archiv. 437, 534–539 (2000).

    Article  CAS  PubMed  Google Scholar 

  108. Dalbeth, N. et al. Mechanisms of bone erosion in gout: a quantitative analysis using plain radiography and computed tomography. Ann. Rheum. Dis. 68, 1290–1295 (2009).

    Article  CAS  PubMed  Google Scholar 

  109. Towiwat, P. et al. Urate crystal deposition and bone erosion in gout: ‘inside-out’ or ‘outside-in’? A dual-energy computed tomography study. Arthritis Res. Ther. 18, 208 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Dalbeth, N. et al. Enhanced osteoclastogenesis in patients with tophaceous gout: urate crystals promote osteoclast development through interactions with stromal cells. Arthritis Rheum. 58, 1854–1865 (2008).

    Article  CAS  PubMed  Google Scholar 

  111. Chhana, A. et al. Monosodium urate monohydrate crystals inhibit osteoblast viability and function: implications for development of bone erosion in gout. Ann. Rheum. Dis. 70, 1684–1691 (2011).

    Article  CAS  PubMed  Google Scholar 

  112. Chhana, A. et al. Monosodium urate crystals reduce osteocyte viability and indirectly promote a shift in osteocyte function towards a proinflammatory and proresorptive state. Arthritis Res. Ther. 20, 208 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Janssens, H. J. et al. A diagnostic rule for acute gouty arthritis in primary care without joint fluid analysis. Arch. Intern. Med. 170, 1120–1126 (2010). This article describes a diagnostic rule for gout in patients presenting with monoarthritis in clinical practice, which has subsequently been validated in a number of different clinical situations.

    Article  PubMed  Google Scholar 

  114. Vazquez-Mellado, J. et al. Intradermal tophi in gout: a case-control study. J. Rheumatol. 26, 136–140 (1999).

    CAS  PubMed  Google Scholar 

  115. El-Zawawy, H. & Mandell, B. F. Crystal-induced arthritides in the elderly: an update. Rheum. Dis. Clin. North Am. 44, 489–499 (2018).

    Article  PubMed  Google Scholar 

  116. Zhang, W. et al. EULAR evidence based recommendations for gout. Part I: Diagnosis. report of a task force of the Standing Committee for International Clinical Studies Including Therapeutics (ESCISIT). Ann. Rheum. Dis. 65, 1301–1311 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Perez-Ruiz, F., Castillo, E., Chinchilla, S. P. & Herrero-Beites, A. M. Clinical manifestations and diagnosis of gout. Rheum. Dis. Clin. North Am. 40, 193–206 (2014).

    Article  PubMed  Google Scholar 

  118. Neogi, T., Krasnokutsky, S. & Pillinger, M. H. Urate and osteoarthritis: evidence for a reciprocal relationship. Joint Bone Spine https://doi.org/10.1016/j.jbspin.2018.11.002 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Forbess, L. J. & Fields, T. R. The broad spectrum of urate crystal deposition: unusual presentations of gouty tophi. Semin. Arthritis Rheum. 42, 146–154 (2012).

    Article  CAS  PubMed  Google Scholar 

  120. Roddy, E., Zhang, W. & Doherty, M. Are joints affected by gout also affected by osteoarthritis? Ann. Rheum. Dis. 66, 1374–1377 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  121. Perez Ruiz, F., Ruiz Lopez, J. & Herrero Beites, A. M. Influence of the natural history of disease on a previous diagnosis in patients with gout [Spanish]. Reumatol. Clin. 5, 248–251 (2009).

    Article  PubMed  Google Scholar 

  122. Taylor, W. J. et al. Performance of classification criteria for gout in early and established disease. Ann. Rheum. Dis. 75, 178–182 (2016).

    Article  PubMed  Google Scholar 

  123. Janssens, H. et al. Performance of the 2015 ACR-EULAR classification criteria for gout in a primary care population presenting with monoarthritis. Rheumatology 56, 1335–1341 (2017).

    Article  PubMed  Google Scholar 

  124. Kienhorst, L. B., Janssens, H. J., Fransen, J. & Janssen, M. The validation of a diagnostic rule for gout without joint fluid analysis: a prospective study. Rheumatology 54, 609–614 (2015).

    Article  PubMed  Google Scholar 

  125. Lee, K. H., Choi, S. T., Lee, S. K., Lee, J. H. & Yoon, B. Y. Application of a novel diagnostic rule in the differential diagnosis between acute gouty arthritis and septic arthritis. J. Korean Med. Sci. 30, 700–704 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  126. Perez-Ruiz, F., Martin, I. & Canteli, B. Ultrasonographic measurement of tophi as an outcome measure for chronic gout. J. Rheumatol. 34, 1888–1893 (2007).

    PubMed  Google Scholar 

  127. Taylor, W. J. et al. Diagnostic arthrocentesis for suspicion of gout is safe and well tolerated. J. Rheumatol. 43, 150–153 (2016).

    Article  PubMed  Google Scholar 

  128. Ankli, B. et al. Calcium pyrophosphate deposition disease: a frequent finding in patients with long-standing erosive gout. Scand. J. Rheumatol. 47, 127–130 (2018).

    Article  CAS  PubMed  Google Scholar 

  129. Filippucci, E. et al. Ultrasound imaging for the rheumatologist. XLVII. Ultrasound of the shoulder in patients with gout and calcium pyrophosphate deposition disease. Clin. Exp. Rheumatol. 31, 659–664 (2013).

    PubMed  Google Scholar 

  130. Reuss-Borst, M. A., Pape, C. A. & Tausche, A. K. Hidden gout- Ultrasound findings in patients with musculo-skeletal problems and hyperuricemia. Springerplus 3, 592 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Choi, H. K. et al. Dual energy computed tomography in tophaceous gout. Ann. Rheum. Dis. 68, 1609–1612 (2009).

    Article  CAS  PubMed  Google Scholar 

  132. Wang, Y., Deng, X., Xu, Y., Ji, L. & Zhang, Z. Detection of uric acid crystal deposition by ultrasonography and dual-energy computed tomography: a cross-sectional study in patients with clinically diagnosed gout. Medicine 97, e12834 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Ogdie, A. et al. Performance of ultrasound in the diagnosis of gout in a multicenter study: comparison with monosodium urate monohydrate crystal analysis as the gold standard. Arthritis Rheumatol. 69, 429–438 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  134. Logan, J. A., Morrison, E. & McGill, P. E. Serum uric acid in acute gout. Ann. Rheum. Dis. 56, 696–697 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Kang, E. H., Lee, E. Y., Lee, Y. J., Song, Y. W. & Lee, E. B. Clinical features and risk factors of postsurgical gout. Ann. Rheum. Dis. 67, 1271–1275 (2008).

    Article  CAS  PubMed  Google Scholar 

  136. Kannangara, D. R. et al. Fractional clearance of urate: validation of measurement in spot-urine samples in healthy subjects and gouty patients. Arthritis Res. Ther. 14, R189 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  137. Perez-Ruiz, F., Hernandez-Baldizon, S., Herrero-Beites, A. M. & Gonzalez-Gay, M. A. Risk factors associated with renal lithiasis during uricosuric treatment of hyperuricemia in patients with gout. Arthritis Care Res. 62, 1299–1305 (2010).

    Article  CAS  Google Scholar 

  138. Stamp, L. & Dalbeth, N. Screening for hyperuricaemia and gout: a perspective and research agenda. Nat. Rev. Rheumatol. 10, 752–756 (2014).

    Article  PubMed  Google Scholar 

  139. Khanna, D. 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. 64, 1431–1446 (2012).

    Article  CAS  Google Scholar 

  140. Richette, P. et al. 2016 updated EULAR evidence-based recommendations for the management of gout. Ann. Rheum. Dis. 76, 29–42 (2017).

    Article  CAS  PubMed  Google Scholar 

  141. Abhishek, A. et al. Monosodium urate monohydrate crystal deposits are common in asymptomatic sons of patients with gout: the Sons of Gout Study. Arthritis Rheumatol. 70, 1847–1852 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Howard, R. G. et al. Reproducibility of musculoskeletal ultrasound for determining monosodium urate deposition: concordance between readers. Arthritis Care Res. 63, 1456–1462 (2011).

    Article  CAS  Google Scholar 

  143. Pineda, C. et al. Joint and tendon subclinical involvement suggestive of gouty arthritis in asymptomatic hyperuricemia: an ultrasound controlled study. Arthritis Res. Ther. 13, R4 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  144. Fisher, M. C., Rai, S. K., Lu, N., Zhang, Y. & Choi, H. K. The unclosing premature mortality gap in gout: a general population-based study. Ann. Rheum. Dis. 76, 1289–1294 (2017).

    Article  PubMed  Google Scholar 

  145. Andres, M. et al. Cardiovascular risk of patients with gout seen at rheumatology clinics following a structured assessment. Ann. Rheum. Dis. 76, 1263–1268 (2017).

    Article  CAS  PubMed  Google Scholar 

  146. Maglio, C. et al. Effects of bariatric surgery on gout incidence in the Swedish Obese Subjects study: a non-randomised, prospective, controlled intervention trial. Ann. Rheum. Dis. 76, 688–693 (2017).

    Article  PubMed  Google Scholar 

  147. Waldman, B. et al. Effect of fenofibrate on uric acid and gout in type 2 diabetes: a post-hoc analysis of the randomised, controlled FIELD study. Lancet Diabetes Endocrinol. 6, 310–318 (2018).

    Article  CAS  PubMed  Google Scholar 

  148. Kimura, K. et al. Febuxostat therapy for patients with stage 3 CKD and asymptomatic hyperuricemia: a randomized trial. Am. J. Kidney Dis. 72, 798–810 (2018).

    Article  CAS  PubMed  Google Scholar 

  149. Khanna, D. et al. 2012 American College of Rheumatology guidelines for management of gout. Part 2: therapy and antiinflammatory prophylaxis of acute gouty arthritis. Arthritis Care Res. 64, 1447–1461 (2012).

    Article  CAS  Google Scholar 

  150. Rubin, B. R. et al. Efficacy and safety profile of treatment with etoricoxib 120 mg once daily compared with indomethacin 50 mg three times daily in acute gout: a randomized controlled trial. Arthritis Rheum. 50, 598–606 (2004).

    Article  CAS  PubMed  Google Scholar 

  151. Janssens, H. J., Janssen, M., van de Lisdonk, E. H., van Riel, P. L. & van Weel, C. Use of oral prednisolone or naproxen for the treatment of gout arthritis: a double-blind, randomised equivalence trial. Lancet 371, 1854–1860 (2008).

    Article  CAS  PubMed  Google Scholar 

  152. Terkeltaub, R. et al. High versus low dosing of oral colchicine for early acute gout flare. Arthritis Rheum. 62, 1060–1068 (2010).

    Article  CAS  PubMed  Google Scholar 

  153. Schlesinger, N. et al. Canakinumab relieves symptoms of acute flares and improves health-related quality of life in patients with difficult to treat gouty arthritis by suppressing inflammation: results of a randomized, dose-ranging study. Arthritis Res. Ther. 13, R53 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Dalbeth, N. et al. Effects of allopurinol dose escalation on bone erosion and urate volume in gout: a dual energy CT imaging study of a randomized controlled trial. Arthritis Rheumatol. https://doi.org/10.1002/art.40929 (2019).

  155. Doherty, M. et al. Efficacy and cost-effectiveness of nurse-led care involving education and engagement of patients and a treat-to-target urate-lowering strategy versus usual care for gout: a randomised controlled trial. Lancet 392, 1403–1412 (2018). This large, randomized controlled trial demonstrated major improvements in clinical outcomes in patients receiving nurse-led gout care according to rheumatology guidelines for gout management compared with usual care by a general practitioner.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Hui, M. et al. The British Society for Rheumatology guideline for the management of gout. Rheumatology 56, 1056–1059 (2017).

    Article  CAS  PubMed  Google Scholar 

  157. Qaseem, A., Harris, R. P. & Forciea, M. A. Management of acute and recurrent gout: a clinical practice guideline from the American College of Physicians. Ann. Intern. Med. 166, 58–68 (2017).

    Article  PubMed  Google Scholar 

  158. Taylor, T. H., Mecchella, J. N., Larson, R. J., Kerin, K. D. & Mackenzie, T. A. Initiation of allopurinol at first medical contact for acute attacks of gout: a randomized clinical trial. Am. J. Med. 125, 1126–1134 (2012). e1127.

    Article  CAS  PubMed  Google Scholar 

  159. Hill, E. M., Sky, K., Sit, M., Collamer, A. & Higgs, J. Does starting allopurinol prolong acute treated gout? A randomized clinical trial. J. Clin. Rheumatol. 21, 120–125 (2015).

    Article  PubMed  Google Scholar 

  160. Yamanaka, H. et al. Stepwise dose increase of febuxostat is comparable with colchicine prophylaxis for the prevention of gout flares during the initial phase of urate-lowering therapy: results from FORTUNE-1, a prospective, multicentre randomised study. Ann. Rheum. Dis. 77, 270–276 (2018). This clinical trial showed that gradual escalation of the febuxostat dose reduces the risk of gout flares compared with full-dose febuxostat when starting urate-lowering therapy; this study also demonstrated the efficacy of low-dose colchicine (0.5 mg daily) in reducing gout flares when initiating urate-lowering therapy.

    Article  CAS  PubMed  Google Scholar 

  161. Becker, M. et al. Febuxostat compared with allopurinol in patients with hyperuricaemia and gout. N. Engl. J. Med. 353, 2450–2461 (2005).

    Article  CAS  PubMed  Google Scholar 

  162. Becker, M., Schumacher, H. R., MacDonald, P., Lloyd, E. & Lademacher, C. Clinical efficacy and safety of successful longterm urate lowering with febuxostat or allopurinol in subjects with gout. J. Rheumatol. 36, 1273–1282 (2009).

    Article  CAS  PubMed  Google Scholar 

  163. Stamp, L. et al. A randomised controlled trial of the efficacy and safety of allopurinol dose escalation to achieve target serum urate in people with gout. Ann, Rheum, Dis. 76, 1522–1528 (2017). This randomized controlled trial showed that allopurinol dose escalation can be used to attain a target serum urate concentration in most patients with gout.

    Article  CAS  Google Scholar 

  164. Stamp, L. et al. Allopurinol dose escalation to achieve serum urate below 6 mg/dl: an open label extension study. Ann. Rheum. Dis. 76, 2065–2070 (2017).

    Article  CAS  PubMed  Google Scholar 

  165. Stamp, L. K. et al. How much allopurinol does it take to get to target urate? Comparison of actual dose with creatinine clearance-based dose. Arthritis Res. Ther. 20, 255 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Wallace, M. C. et al. Association between ABCG2 rs2231142 and poor response to allopurinol: replication and meta-analysis. Rheumatology 57, 656–660 (2018).

    Article  CAS  PubMed  Google Scholar 

  167. Stamp, L., Day, R. & Yun, J. Allopurinol hypersensitivity: investigating the cause and minimizing the risk. Nat. Rev. Rheumatol. 12, 235–242 (2016).

    Article  CAS  PubMed  Google Scholar 

  168. Hung, S. I. et al. HLA-B*5801 allele as a genetic marker for severe cutaneous adverse reactions caused by allopurinol. Proc. Natl Acad. Sci. USA 102, 4134–4139 (2005). This was the original description of HLA-B*5801 as a genetic marker for allopurinol-induced SCAR.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Ko, T. M. et al. Use of HLA-B*58:01 genotyping to prevent allopurinol induced severe cutaneous adverse reactions in Taiwan: national prospective cohort study. BMJ 351, h4848 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  170. Chung, W.-H. et al. Insights into the poor prognosis of allopurinol-induced severe cutaneous adverse reactions: the impact of renal insufficiency, high plasma levels of oxypurinol and granulysin. Ann. Rheum. Dis. 74, 2157–2164 (2015).

    Article  CAS  PubMed  Google Scholar 

  171. Shibagaki, Y., Ohno, I., Hosoya, T. & Kimura, K. Safety, efficacy and renal effect of febuxostat in patients with moderate-to-severe kidney dysfunction. Hypertens. Res. 37, 919–925 (2014).

    Article  CAS  PubMed  Google Scholar 

  172. Saag, K. et al. Impact of febuxostat on renal function in gout subjects with moderate-to-severe renal impairment. Arthritis Rheum. 68, 2035–2043 (2016).

    Article  CAS  Google Scholar 

  173. White, W. et al. Cardiovascular safety of febuxostat or allopurinol in patients with gout. N. Engl. J. Med. 378, 1200–1210 (2018). The published results of the large, randomized controlled CARES trial show increased cardiovascular and all-cause mortality from febuxostat treatment compared with allopurinol treatment in individuals with gout and established cardiovascular disease.

    Article  CAS  PubMed  Google Scholar 

  174. Choi, H., Neogi, T., Stamp, L., Dalbeth, N. & Terkeltaub, R. Implications of the cardiovascular safety of febuxostat and allopurinol in patients with gout and cardiovascular morbidities (CARES) trial and associated FDA public safety alert. Arthritis Rheumatol. 70, 1702–1709 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Pui, K., Gow, P. & Dalbeth, N. Efficacy and tolerability of probenecid as urate-lowering therapy in gout; clinical experience in high-prevalence population. J. Rheumatol. 40, 872–876 (2013).

    Article  CAS  PubMed  Google Scholar 

  176. Bardin, T. et al. Lesinurad in combination with allopurinol: a randomised, double-blind, placebo-controlled study in patients with gout with inadequate response to standard of care (the multinational CLEAR 2 study). Ann. Rheum. Dis. 76, 811–820 (2016).

    Article  PubMed  CAS  Google Scholar 

  177. Saag, K. et al. Lesinurad combined with allopurinol: randomized, double-blind, placebo-controlled study in gout subjects with inadequate response to standard of care allopurinol (a US-based study). Arthritis Rheum. 69, 203–212 (2017).

    Article  CAS  Google Scholar 

  178. Dalbeth, N. et al. Lesinurad, a selective uric acid reabsorption inhibitor, in combination with febuxostat in patients with tophaceous gout. Arthritis Rheum. 69, 1903–1913 (2017).

    Article  CAS  Google Scholar 

  179. Sundy, J. et al. Efficacy and tolerability of pegloticase for the treatment of chronic gout in patients refractory to conventional treatment: two randomized controlled trials. JAMA 306, 711–720 (2011).

    Article  CAS  PubMed  Google Scholar 

  180. Becker, M. et al. Long-term safety of pegloticase in chronic gout refractory to conventional treatment. Ann. Rheum. Dis. 72, 1469–1474 (2013).

    Article  CAS  PubMed  Google Scholar 

  181. Lipsky, P. et al. Pegloticase immunogenicity: the relationship between efficacy and antibody development in patients treated for refractory chronic gout. Arthritis Res. Ther. 16, R60 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Becker, M. et al. The urate-lowering efficacy and safety of febuxostat in the treatment of the hyperuricaemia of gout: the CONFIRMS trial. Arthritis Res. Ther. 12, R63 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. Li, X. et al. Serum uric acid levels and multiple health outcomes: umbrella review of evidence from observational studies, randomised controlled trials, and Mendelian randomisation studies. BMJ 357, j2376 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  184. Terkeltaub, R., Furst, D., DiGiacinto, J., Kook, K. & Davis, M. Novel evidence-based colchicine dose-reduction algorithm to predict and prevent colchicine toxicity in the presence of cytochrome P450 3A4/P-glycoprotein inhibitors. Arthritis Rheum. 63, 2226–2237 (2011).

    Article  CAS  PubMed  Google Scholar 

  185. Jeyaruban, A., Soden, M. & Larkins, S. General practitioners’ perspectives on the management of gout: a qualitative study. Postgrad. Med. J. 92, 603–607 (2016).

    Article  PubMed  Google Scholar 

  186. Nielsen, S. et al. Weight loss for overweight and obese individuals with gout: a systematic review of longitudinal studies. Ann. Rheum. Dis. 76, 1870–1882 (2017).

    Article  PubMed  Google Scholar 

  187. Holland, R. & McGill, N. Comprehensive dietary education in treated gout patients does not further improve serum urate. Int. Med. J. 45, 189–189 (2015).

    Article  CAS  Google Scholar 

  188. Becker, M. A. et al. Quality of life and disability in patients with treatment-failure gout. J. Rheumatol. 36, 1041–1048 (2009).

    Article  PubMed  Google Scholar 

  189. Kleinman, N. L. et al. The impact of gout on work absence and productivity. Value Health 10, 231–237 (2007).

    Article  PubMed  Google Scholar 

  190. Khanna, P. P. et al. Tophi and frequent gout flares are associated with impairments to quality of life, productivity, and increased healthcare resource use: results from a cross-sectional survey. Health Qual. Life Outcomes 10, 117 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  191. Chandratre, P. et al. Health-related quality of life in gout in primary care: baseline findings from a cohort study. Semin. Arthritis Rheum. 48, 61–69 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  192. Chandratre, P. et al. Health-related quality of life in gout: a systematic review. Rheumatology 52, 2031–2040 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Khanna, P. P. et al. Health-related quality of life and treatment satisfaction in patients with gout: results from a cross-sectional study in a managed care setting. Patient Prefer. Adherence 9, 971–981 (2015).

    PubMed  PubMed Central  Google Scholar 

  194. Shields, G. E. & Beard, S. M. A systematic review of the economic and humanistic burden of gout. Pharmacoeconomics 33, 1029–1047 (2015).

    Article  PubMed  Google Scholar 

  195. Wertheimer, A., Morlock, R. & Becker, M. A. A revised estimate of the burden of illness of gout. Curr. Ther. Res. Clin. Exp. 75, 1–4 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  196. Flores, N. M., Nuevo, J., Klein, A. B., Baumgartner, S. & Morlock, R. The economic burden of uncontrolled gout: how controlling gout reduces cost. J. Med. Econ. 22, 1–6 (2018).

    Article  PubMed  Google Scholar 

  197. Strand, V., Khanna, D., Singh, J. A., Forsythe, A. & Edwards, N. L. Improved health-related quality of life and physical function in patients with refractory chronic gout following treatment with pegloticase: evidence from phase III randomized controlled trials. J. Rheumatol. 39, 1450–1457 (2012).

    Article  CAS  PubMed  Google Scholar 

  198. Dalbeth, N. et al. Effects of skim milk powder enriched with glycomacropeptide and G600 milk fat extract on frequency of gout flares: a proof-of-concept randomised controlled trial. Ann. Rheum. Dis. 71, 929–934 (2012).

    Article  CAS  PubMed  Google Scholar 

  199. Stamp, L. et al. Clinically insignificant effect of supplemental vitamin C on serum urate in patients with gout; a pilot randomised controlled trial. Arthritis Rheum. 65, 1636–1642 (2013).

    Article  CAS  PubMed  Google Scholar 

  200. Rai, S. K. et al. Key barriers to gout care: a systematic review and thematic synthesis of qualitative studies. Rheumatology 57, 1282–1292 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  201. Robinson, P. C., Taylor, W. J. & Dalbeth, N. An observational study of gout prevalence and quality of care in a national Australian general practice population. J. Rheumatol. 42, 1702–1707 (2015).

    Article  CAS  PubMed  Google Scholar 

  202. FitzGerald, J. D. et al. Development of the American College of Rheumatology electronic clinical quality measures for gout. Arthritis Care Res. 70, 659–671 (2018).

    Article  Google Scholar 

  203. Roddy, E., Zhang, W. & Doherty, M. Concordance of the management of chronic gout in a UK primary-care population with the EULAR gout recommendations. Ann. Rheum. Dis. 66, 1311–1315 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Scheepers, L. et al. Medication adherence among patients with gout: a systematic review and meta-analysis. Semin. Arthritis Rheum. 47, 689–702 (2018).

    Article  PubMed  Google Scholar 

  205. Kuo, C. F., Grainge, M. J., Mallen, C., Zhang, W. & Doherty, M. Eligibility for and prescription of urate-lowering treatment in patients with incident gout in England. JAMA 312, 2684–2686 (2014).

    Article  PubMed  Google Scholar 

  206. Duyck, S. D., Petrie, K. J. & Dalbeth, N. “You don’t have to be a drinker to get gout, but it helps”: a content analysis of the depiction of gout in popular newspapers. Arthritis Care Res. 68, 1721–1725 (2016).

    Article  Google Scholar 

  207. Bulbin, D. et al. Improved gout outcomes in primary care using a novel disease management program: a pilot study. Arthritis Care Res. 70, 1679–1685 (2018).

    Article  CAS  Google Scholar 

  208. Goldfien, R., Pressman, A., Jacobson, A., Ng, M. & Avins, A. A pharmacist-staffed, virtual gout management clinic for achieving target serum uric acid levels: a randomized clinical trial. Perm. J. 20, 18–23 (2016).

    Google Scholar 

  209. Mikuls, T. R. et al. Adherence and outcomes with urate-lowering therapy: a site-randomized trial. Am. J. Med. 132, 354–361 (2018).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  210. Reginato, A. M., Mount, D. B., Yang, I. & Choi, H. K. The genetics of hyperuricaemia and gout. Nat. Rev. Rheumatol. 8, 610–621 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors are grateful to A. Nakayama for editing assistance. H.K.C. is supported by the US National Institutes of Health (AR060772).

Author information

Authors and Affiliations

Authors

Contributions

Introduction (N.D.); Epidemiology (H.K.C.); Mechanisms/pathophysiology (H.M. and L.A.B.J.); Diagnosis, screening and prevention (F.P.-R. and N.D.); Management (L.K.S.); Quality of life (P.P.K.); Outlook (N.D.); overview of the Primer (N.D.).

Corresponding author

Correspondence to Nicola Dalbeth.

Ethics declarations

Competing interests

N.D. has received speaking fees from Pfizer, Horizon, Janssen and AbbVie; consulting fees from Horizon, Hengrui and Kowa; and research funding from Amgen and AstraZeneca; and is currently principal investigator on a clinical trial of intensive urate-lowering therapy (funded by the Health Research Council of New Zealand). Within the past 5 years, N.D. has been principal investigator on a clinical trial of febuxostat in early gout, and received consulting or speaking fees from Takeda, Menarini and Teijin. H.K.C. received research support from AstraZeneca and consulting fees from Takeda, Selecta, GSK and Horizon. L.A.B.J. has received a speaking fee from Novartis, research funding from Ardea Biosciences/AstraZeneca, and is Scientific Advisory Board member of Olatec Therapeutics LLC. P.P.K. has received research grants from Ironwood, Sobi and Horizon. F.P.-R. has been an advisor for Amgen, Horizon, Grünenthal, Menarini, Sanofi and Syneos-Health; a speaker for Amgen, Astellas, Grünenthal, Lilly, Logarithm, Menarini and the Spanish Foundation for Rheumatology; received investigation grants from Cruces Rheumatology Association and the Spanish Foundation for Rheumatology; and received congress funding from Lilly, Novartis and the European League Against Rheumatism (EULAR). L.K.S. has received speaking fees from Amgen and is currently principal investigator on a clinical trial of colchicine prophylaxis (funded by the Health Research Council of New Zealand). L.K.S. is a member of the Medicines Adverse Reaction Committee of New Zealand and the New Zealand Pharmaceutical Management Agency (PHARMAC) Rheumatology Subcommittee. H.M. declares no competing interests.

Additional information

Peer review information

Nature Reviews Disease Primers thanks J.G. Puig, M. Pillinger, P. Richette, M. Andrés and R. Burgos-Vargas for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dalbeth, N., Choi, H.K., Joosten, L.A.B. et al. Gout. Nat Rev Dis Primers 5, 69 (2019). https://doi.org/10.1038/s41572-019-0115-y

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41572-019-0115-y

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing