Next Article in Journal
Safety and Efficacy of the Nit-Occlud® Coil for Percutaneous Closure of Various Sizes of PDA
Previous Article in Journal
The Associations of Mental Health Disturbances, Self-Esteem, and Partner Violence Victimization with Condom Use in Spanish Adolescents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Efficacy of Xanthine Oxidase Inhibitors in Lowering Serum Uric Acid in Chronic Kidney Disease: A Systematic Review and Meta-Analysis

1
Molecular Genetic Epidemiology Section, Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
2
Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
3
Department of Internal Medicine, The Hospital of Central Connecticut, New Britain, CT 06052, USA
4
Howard University College of Medicine, Washington, DC 20059, USA
5
Biostatistics and Clinical Epidemiology Service, National Institutes of Health, Bethesda, MD 20814, USA
6
Department of Internal Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
7
Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20814, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2022, 11(9), 2468; https://doi.org/10.3390/jcm11092468
Submission received: 15 March 2022 / Revised: 18 April 2022 / Accepted: 20 April 2022 / Published: 27 April 2022
(This article belongs to the Section Nephrology & Urology)

Abstract

:
Objective: Current guidelines for gout recommend a treat-to-target approach with serum uric acid (SUA). However, there is little evidence for the dose-dependent effects of urate-lowering therapy (ULT). Herein, we analyzed the reported SUA-lowering effect and SUA target achievement differences for various doses of xanthine oxidase inhibitors. Methods: Approved ULT drugs were selected from the FDA Drug Database. We included prospective randomized controlled trials of ULT drugs from ClinicalTrials.gov, articles published in the journal “Drugs”, and Embase, a literature database. A meta-analysis was performed to determine the ability of different ULT drugs and doses to lower and maintain a target SUA < 6 mg/dL. Results: We identified 35 trials including 8172 patients with a baseline SUA of 8.92 mg/dL. The allopurinol, febuxostat, and topiroxostat showed dose-proportional SUA-lowering responses. Compared with allopurinol 300 mg daily, febuxostat 80 mg daily and 120 mg daily more effectively maintained SUA < 6 mg/dL. Conclusion: Allopurinol, febuxostat, and topiroxostat showed dose-proportional ability to lower and achieve a target SUA < 6 mg/dL. Significance and Innovations. We showed dose-dependent SUA lowering effects of allopurinol, febuxostat, and topiroxostat. Febuxostat is effective at ULT compared to allopurinol and could be potentially offered as an alternative agent when patients (1) have CKD, (2) have the human leukocyte antigen HLA-B*5801 allele, and (3) become refractory to allopurinol. Gradual allopurinol dose increase with a lower starting dose is needed in CKD.

1. Introduction

Uric acid, the final metabolic product of purine degradation, is a known or potential biomarker for various pathological conditions. Hyperuricemia is defined as a serum uric acid (SUA) level of >6.8 mg/dL or 404.5 µmol/L at physiological pH [1] and can lead to the development of gout and nephrolithiasis. It also has been associated with insulin resistance, type 2 diabetes mellitus (T2DM), hypertension, chronic kidney disease (CKD), cardiovascular disease, and tumor lysis syndrome [1,2,3,4,5,6,7,8,9,10,11,12,13]. Obesity is a strong, modifiable risk factor for gout and hyperuricemia [14]. Increasing evidence suggests that insulin resistance is a key mediator of the association between obesity and hyperuricemia [10,13,14].
Gout occurs when highly immunogenic monosodium urate crystals [15] form in joints and soft tissue and is promoted when the SUA concentrations exceed solubility limits. These crystals produce painful attacks and debilitating bone erosion [16]. According to the 2015–2016 National Health and Nutrition Examination Survey, the prevalence of hyperuricemia and gout in the United States was 20.1% and 3.9%, respectively, and both increased in prevalence since the previous survey in 1988–1994 [17,18]. An increasing prevalence of gout has also been reported in other developed countries [19].
Urate-lowering therapy (ULT) is the mainstay of chronic gout management. Both the 2020 American College of Rheumatology (ACR) and 2016 European Alliance of Associations for Rheumatology (EULAR) guidelines recommend a treat-to-target management strategy in the absence of tophi (aggregates of monosodium urate crystals), aiming to reduce SUA to <6 mg/dL. EULAR further recommends a target of SUA of <5 mg/dL when tophi are present [20,21]. As treatment success is inversely related to baseline SUA [22], researchers should account for baseline SUA when examining the effectiveness of ULT, as variations in baseline SUA can result in different efficacies of ULT.
ULTs comprise xanthine oxidase inhibitors (XOIs), uricosuric agents, and recombinant uricase. The XOIs inhibit the enzyme xanthine oxidase that metabolizes hypoxanthine to uric acid and thereby reduces uric acid generation. The most commonly used XOI, allopurinol, is an analog of hypoxanthine and xanthine; it competes with these molecules in binding xanthine oxidase and inhibits the formation of uric acid [23]. Febuxostat non-selectively inhibits oxidized and reduced forms of xanthine oxidase [23]. It is a competitive inhibitor of the enzyme. Topiroxostat, a selective non-purine molecule, competitively inhibits xanthine oxidase [24]. Allopurinol and febuxostat are approved for use in the United States, whereas topiroxostat is only approved in Japan. The 2020 ACR and 2016 EULAR guidelines recommend low-dose allopurinol as a first-line treatment for gout [20,21]. Although febuxostat has better SUA-lowering efficacy than allopurinol, there are ongoing concerns regarding cardiovascular safety arising from the Cardiovascular Safety of Febuxostat and Allopurinol in Patients With Gout study [25]. However, more recent studies show no increased cardiovascular risk after febuxostat initiation compared with allopurinol [26,27].
Uricosuric drugs, unlike XOIs, prevent the reuptake of uric acid by the proximal convoluted tubule, primarily through inhibition of the human urate-1 transporter (URAT1). Uricosuric agents include probenecid, benzbromarone, and lesinurad.
Recombinant uricases convert uric acid into a more water-soluble compound, allantoin, for excretion in the urine. Pegloticase and rasburicase are recombinant uricases. Both are pegylated to increase plasma half-life by reducing renal excretion, and pegylation may also provide resistance to proteolysis and reduce antigenicity. Both agents are recommended for treatment-refractory gout based on their efficacy in lowering SUA and promoting the resolution of tophi [28].
Understanding the ability of ULT drugs to lower and maintain a target SUA is critical for clinicians to manage and treat hyperuricemia and gout appropriately. Several studies have demonstrated the efficacy of different ULT drugs across class and dose. Here we performed a systematic review and meta-analysis to determine the SUA-lowering effect and achievement difference (AD) in maintaining the target SUA for different ULT drugs at various doses, considering baseline SUA in hyperuricemic patients.

2. Materials and Methods

2.1. Data Collection Strategy

We reviewed approved drugs for hyperuricosemia listed in the United States Food and Drug Administration’s Approved Drug Product Database (https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm, accessed 14 June 2019). We selected allopurinol, febuxostat, topiroxostat, benzbromarone, probenecid, lesinurad, rasburicase, and pegloticase for inclusion in this systematic review.
We included randomized prospective clinical trials (phase 1 to 4) of ULT drugs for the treatment of gout, hyperuricemia, or tumor lysis syndrome, regardless of the study masking method. Trials that involved dose titration intervals in step-up protocols were included by recording the extent of SUA-lowering after participants reached the maintenance dose for the time defined by each study protocol.
Records were collected from the following four sources (accessed 14 June 2019): ClinicalTrials.gov (accessed 14 June 2019, reviewed: n = 300 studies; included: n = 27), articles published in the journal Drugs (ISSN: 0012-6667) (reviewed: n = 236 studies; included: n = 19), Embase (embase.com, accessed 14 June 2019, reviewed: n = 732 studies; included: n = 34), and one publication [29] that was a reference in an included study. We used the following search terms: allopurinol, febuxostat, topiroxostat, gout, and hyperuricemia. The studies were included up to June 2019. In total, we included 81 studies.

2.2. Data Extraction and Imputation of Missing Values

Data were extracted from the journal article associated with each clinical trial or the ClinicalTrials.gov entry when a journal article was unavailable. Data were entered into a standardized data extraction form.
In cases of missing data, we calculated the percent SUA change from initial and final SUA levels ( FinalSUA InitialSUA InitialSUA × 100 ), computed standard error (SE) if standard deviation (SD) was available ( SE = SD n ), and estimated SD if SE or 95% confidence intervals (CIs) were provided ( SD = n × Upper 95 % CI Lower 95 % CI 3.92 ).
SD and SE were unavailable for 34 out of 42 (80.95%) allopurinol arms, 13 out of 34 (38.24%) febuxostat arms, 11 out of 21 (52%) topiroxostat arms, 15 out of 16 (94%) benzbromarone arms, and 3 out of 4 (75%) probenecid arms. For these studies, we estimated the drug-specific pooled variance of the SE ( i = 1 n ( SE 1 2 n 1 + SE 2 2 n 2 + + SE n 2 n n ) and imputed that value as the SE for each trial arm.

2.3. Quality Control Using Inclusion/Exclusion Criteria

2.3.1. SUA-Lowering Analysis

In the SUA-lowering analysis, we excluded trial arms for any of the following six reasons: (1) SD or SE for percent SUA lowering were not reported, which are required for meta-regression; (2) we excluded placebo arms; (3) baseline SUA was within normal range (<6.8 mg/dL); (4) a dosing frequency other than once daily was used; (5) a dose titration scheme was used; or (6) the study did not define CKD.
After excluding 152 trial arms (64.68%; Figure 1 and Supplemental Table S1), 35 studies encompassing the following 83 trial arms were included: 29 arms for allopurinol, 4 arms for benzbromarone, 28 arms for febuxostat, 1 arm for lesinurad, 1 arm for probenecid, 16 arms for topiroxostat, and 4 arms for a combination of two ULT drugs (Supplemental Table S1). Only allopurinol, febuxostat, and topiroxostat were included in the final meta-analysis due to insufficient data for other ULT drugs.

2.3.2. Achievement Difference Analysis

In the AD analysis, we selected trial arms based on 2020 ACR and 2016 EULAR guidelines [20,21]. As the 2020 ACR guidelines strongly recommend allopurinol as a first-line ULT, we included all trials using allopurinol 300 mg daily (the standard dose) as a control group [20]. To analyze studies targeting maintenance of SUA <6 mg/dL, we included the following 7 trial arms for febuxostat: 2 arms at 40 mg daily, 4 arms at 80 mg daily, and 1 arm at 120 mg daily.

3. Statistical Analysis

Previous studies of ULT drugs used the following different dosing frequencies: daily, twice daily, three times daily, or every four days. We chose to focus on daily doses, the most commonly used regimen, to harmonize the SUA-lowering effect of different ULT drugs. Thus, we excluded all other dosing frequencies.
In estimation of the dose-dependent SUA-lowering effect, we conducted a meta-analysis using the metareg command of Stata, version 16 (StataCorp; 2019, College Station, TX, USA: StataCorp LLC.). To account for the effect of baseline SUA on dose-dependent SUA-lowering outcomes, we selected only trials including subjects with an elevated baseline SUA of >6 mg/dL. We adjusted the analysis for covariates, including CKD diagnosis, sample size, length of follow-up, collection site, and proportion of male participants in each arm, as all studies included a preponderance of males. The proportion of male participants was transformed into a binary variable based on the following median values for trial arms (0.82 for allopurinol arms, 0.95 for febuxostat arms, and 0.91 for topiroxostat arms): 1 for more male-dominant arms and 0 for less male-dominant arms. This transformation was performed because the proportion of male participants was highly skewed, and this degraded model fit when it was introduced as a covariate. The meta-regression was performed only for allopurinol, febuxostat, and topiroxostat after excluding unqualified trial arms.
To compare AD among ULT drugs, we conducted a meta-analysis using the meta command of Stata, version 16 (StataCorp; 2019, College Station, TX, USA: StataCorp LLC.) model. For the analysis, we selected only arms, including subjects with a baseline SUA >6.8 mg/dL, which we defined as the upper limit of normal. In the model, we entered the numbers of participants who achieved and did not achieve the intended therapeutic outcome, which yielded the efficacy of the intended therapeutic effect. Systematic bias and heterogeneity among studies are shown as a funnel plot (Supplemental Figure S4).

4. Results

4.1. SUA-Lowering Analysis

4.1.1. Trial Characteristics

The SUA-lowering meta-analysis included 8172 patients with a baseline SUA of 8.92 ± 0.77 mg/dL (mean ± SD; range, 6.88–10.40 mg/dL) after imputation. There were 3465 patients receiving allopurinol with a baseline SUA of 8.82 ± 0.74 mg/dL, 3899 patients on febuxostat with a baseline SUA of 9.25 ± 0.72 mg/dL, 506 patients on topiroxostat with a baseline SUA of 8.66 ± 0.55 mg/dL, 93 patients on benzbromarone with baseline SUA of 8.74 ± 0.34 mg/dL, 35 patients on probenecid with a baseline SUA of 9.07 ± 1.2 mg/dL, and 67 patients on a combination of ULT drugs with a baseline SUA of 8.35 ± 1.75 mg/dL.

4.1.2. Meta-Regression

We conducted random-effects meta-regression to estimate the extent of SUA-lowering based on the dose of three xanthine oxidase inhibitors, allopurinol, febuxostat, and topiroxostat. Allopurinol (n = 29) dose was positively related to the extent of SUA-lowering (β = 0.05, SE = 0.01, p < 0.001, 95% CI 0.036, 0.072) (Table 1 and Supplement Figure S1). In the covariate model (n = 19), the extent of SUA-lowering was significantly related to allopurinol dose (β = 0.36, SE = 0.07, p = 0.001, 95% CI 0.19, 0.52) and marginally significantly related to CKD diagnosis (β = 27.62, SE = 12.71, p = 0.062, 95% CI −1.698, 56.942) (Table 2).
Febuxostat (n = 28) dose was positively related to the extent of serum uric acid-lowering (β = 0.18, SE = 0.03, p < 0.001, 95% CI 0.124, 0.230) (Table 1 and Supplemental Figure S2). The covariate model (n = 25) also showed that the extent of SUA-lowering was significantly related to febuxostat dose (β = 0.23, SE = 0.04, p < 0.001, 95% CI 0.151, 0.301) (Table 3).
The Topiroxostat (n = 16) dose was also positively related to the extent of SUA-lowering (β = 0.10, SE = 0.02, p < 0.001, 95% CI 0.059, 0.141) (Table 1 and Supplement Figure S3). In the covariate model (n = 16), the extent of SUA-lowering was significantly related to the topiroxostat dose (β = 0.13, SE = 0.04, p = 0.007, 95% CI 0.044, 0.220) and proportion of male participants (β = −14.22, SE = 5.16, p = 0.020, 95% CI −25.720, −2.713) (Table 4). Furthermore, a post-hoc analysis showed that the topiroxostat dose was marginally significantly related to the extent of SUA-lowering among trial arms with a greater male predominance (n = 8, β = 0.11, SE = 0.04, p = 0.058, 95% CI −0.006, 0.217) but not among trial arms with a lesser male predominance (n = 8, β = 0.03, SE = 0.07, p = 0.581, 95% CI −0.171, 0.241).

4.2. Achievement Difference Analysis

4.2.1. Trial Characteristics

We used achievement difference (AD) meta-analysis to examine which study arms achieved the target serum uric acid level, as defined in each study. This analysis included 1849 patients with a baseline SUA of 9.79 ± 0.18 mg/dL (mean ± SD; range, 9.43–9.98 mg/dL). There were 632 patients on allopurinol with a baseline SUA of 9.83 ± 0.13 mg/dL and 1217 patients on febuxostat with a baseline SUA of 9.76 ± 0.21 mg/dL.

4.2.2. Meta-Analysis

Two trial arms with febuxostat 40 mg did not show a significant AD in maintaining SUA < 6 mg/dL when compared with allopurinol 300 mg daily (AD = 0.05, 95% CI −0.018, 0.109) (Table 5 and Figure 2). However, four arms with febuxostat 80 mg (AD = 0.25, 95% CI 0.165, 0.325) and one arm with febuxostat 120 mg (AD = 0.40, 95% CI 0.326, 0.483) showed significant AD outcomes compared with allopurinol 300 mg daily.

5. Discussion

Increased uric acid levels play a central role in the pathogenesis of particular diseases, including gout and tumor lysis syndrome, and are associated with a higher risk of cardiovascular disease [8,9], CKD [11], diabetes mellitus [10], and nonalcoholic fatty liver disease [10]. However, there have been few systematic reviews and meta-analyses of ULT. The present meta-analysis provides insight into the dose effects of different ULT drugs and may help guide clinicians in selecting the appropriate dosing to achieve a specific SUA-lowering effect.
We analyzed the following three different XOIs that showed dose-dependent SUA-lowering effects: allopurinol, febuxostat, and topiroxostat. These agents showed meaningful SUA-lowering effects that persisted after controlling for the presence of CKD, sample size, length of follow-up, collection site, and proportion of male participants. With regards to CKD, concerns have arisen with regard to using allopurinol in CKD due to its potentially detrimental effect on renal function, and untreated hyperuricemia may promote poor outcomes of gout or hyperuricemia [65]. Stamp et al. [66,67] studies were supported by results showing allopurinol dose escalation in CKD can achieve effective SUA with comparable safety; a lower starting dosing was required to achieve target SUA in CrCl < 30 mL/min. The analysis presented here shows CKD has a marginal influence on the SUA-lowering effects of allopurinol and hence supports the current ACR 2020 practice guidelines, which recommend a lower allopurinol starting dose in CKD [20].
The present meta-analysis showed that febuxostat had better SUA-lowering ability than allopurinol or topiroxostat (Table 1). Based on the null model, the expected SUA-lowering effect of allopurinol 300 mg was a 15% reduction from the baseline SUA level. Based on our SUA-lowering analysis, an equivalent outcome would be achieved by febuxostat 83.33 mg or topiroxostat 150 mg (Table 2, Table 3 and Table 4). Previous meta-analyses that examined the comparative efficacy of ULT drugs showed similar AD results as in the present study. Ye et al. [68] report that the efficacy of febuxostat 40 mg daily is higher than that of allopurinol 100–300 mg daily and is greater with higher doses (80–120 mg daily). Borghi et al. [69] showed that doses of febuxostat >40 mg daily are superior to allopurinol at 300 mg daily. Furthermore, Li et al. [70] suggested that febuxostat has greater efficacy at 120 mg daily compared to allopurinol at 300 mg daily. Based on our AD analysis results (Table 5), febuxostat 80 mg daily may be a better treatment option than allopurinol for patients with refractory SUA levels, consistent with 2016 EULAR and 2020 ACR recommendations [20,21].
The meta-analysis presented here suggests that febuxostat could be preferable to allopurinol for treating gout. Given the higher cost and black box warning associated with febuxostat, allopurinol is currently recommended as the first-line treatment for most gout patients. However, studies have shown the cost-effectiveness of febuxostat compared with allopurinol in chronic gout management [71,72]. Thus, it may be worthwhile to further examine the advantages of febuxostat from an economic perspective. Moreover, recent studies show no increased cardiovascular risk of febuxostat compared with allopurinol [26,27]. Apart from its overall greater efficacy, febuxostat may also be a better ULT option in specific instances. First, febuxostat may be more beneficial to patients with mild to moderate renal impairment (creatinine clearance of 30–80 mL/min/1.73 m2), as it is not influenced by renal function [73]. Second, febuxostat may be better for treating patients with the human leukocyte antigen HLA-B*5801 allele [74,75]. Previous studies reveal that allopurinol can cause a spectrum of severe skin reactions, including Stevens-Johnson syndrome and toxic epidermal necrolysis, in patients with the HLA-B*5801 allele [74,75]. Although rare, allopurinol hypersensitivity syndrome can cause serious mortality and morbidity. Therefore, according to ACR 2020 gout guidelines, testing for the HLA–B*5801 allele prior to starting allopurinol is recommended for patients of Southeast Asian descent and for African American patients [20]. Third, febuxostat may be a better choice for patients who are refractory to allopurinol. Individuals with a minor allele (rs2231142) of the ATP-binding cassette ABCG2 transporter show minimal response to allopurinol. Therefore, patients with a higher risk of treatment failure and a lack of biological sensitivity to allopurinol could consider initiating treatment with febuxostat [76].
Strengths of the present study include the analysis of SUA-lowering and AD efficacy of ULT drugs at different doses compared with allopurinol 300 mg daily, which is most commonly used in the United States [77]. We also included trials from various geographic regions, including the United States, Europe, China, Japan, Iran, and India. However, this analysis also has limitations, including (1) the specific details of the studies were not captured due to the nature of the meta-analysis technique; (2) heterogeneity among studies may have resulted from the inclusion of older studies between the 1970s and 1980s, which may have included patients with different lifestyles and employed different trial designs; (3) treatment duration and patient characteristics varied among trials; (4) we did not analyze the potential influence of renal function on treatment efficacy by accounting for patients’ body mass index, sex, or ethnicity; (5) there was a lack of standardized reporting of SUA-lowering effects among included trials.
We recommend that future studies analyze sex differences in hyperuricemia. Although men, compared to women, have a higher risk of developing gout (5.2% vs. 2.7%) and hyperuricemia (20.2% vs. 4.2%) [18], there is evidence that during the late menopausal transition stage, SUA rises in women to levels comparable to those of men at a similar age [78]. In addition, although body mass index is typically considered in clinical trials, future trials involving agents that affect SUA should report detailed information on the body mass index of participants, so that clinicians can better understand appropriate doses of ULT. Furthermore, standardized approaches to reporting data should be considered in future studies to improve the feasibility of subsequent meta-analyses.
In summary, the meta-analysis presented here shows the dose-dependent SUA-lowering effects of allopurinol, febuxostat, and topiroxostat. Further, it suggests that febuxostat is a more effective ULT than allopurinol, especially at higher doses. These findings support the ULT recommendations in the 2016 EULAR and 2020 ACR guidelines.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm11092468/s1, Figure S1: Forest plot of SUA-lowering effect of allopurinol [42,43,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63]; Figure S2: Forest plot of SUA-lowering effect of febuxostat [42,43,45,46,47,51,64,65,66,67,68,69,70]; Figure S3: Forest plot of SUA-lowering effect of topiroxostat [49,50,63,71,72,73,74]; Figure S4: Funnel plot of AD for febuxostat monotherapy vs. allopurinol 300 mg daily [42,43,44,45]; Table S1: Exclusion of the Trial Arms in the Urate-Lowering Therapy Drug Analysis.

Funding

This work was supported by the new faculty research fund of Ajou University School of Medicine.

Data Availability Statement

Raw data are available at https://github.com/fatelist82/Gout-Therapeutics (accessed on 14 March 2022).

Acknowledgments

We appreciate Young Sup Cho and Hyekyung Son for their helpful advice. This work was supported by the new faculty research fund of Ajou University School of Medicine. Financial support was received from the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Education (NRF-2016R1A6A3A11933380) and the Korea Health Technology R&D Project through the Korea Health Industry Development Institute funded by the Ministry of Health & Welfare, Republic of Korea (HI17C2372). Kopp is supported by the NIDDK Intramural Research Program, National Institutes of Health.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Terkeltaub, R. Update on gout: New therapeutic strategies and options. Nat. Rev. Rheumatol. 2010, 6, 30–38. [Google Scholar] [CrossRef] [PubMed]
  2. Anker, S.D.; Doehner, W.; Rauchhaus, M.; Knosalla, C.; Hetzer, R.; Coats, A.J. Uric acid and survival in chronic heart failure: Validation and application in metabolic, functional, and hemodynamic staging. Circulation 2003, 107, 1991–1997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kivity, S.; Kopel, E.; Maor, E.; Abu-Bachar, F.; Segev, S.; Sidi, Y.; Olchovsky, D. Association of Serum Uric Acid and Cardiovascular Disease in Healthy Adults. Am. J. Cardiol. 2013, 111, 1146–1151. [Google Scholar] [CrossRef] [PubMed]
  4. Reynolds, T. Serum uric acid, the endothelium and hypertension: An association revisited. J. Hum. Hypertens. 2007, 21, 591–593. [Google Scholar] [CrossRef] [PubMed]
  5. Tsai, C.-W.; Lin, S.-Y.; Kuo, C.-C.; Huang, C.-C. Serum Uric Acid and Progression of Kidney Disease: A Longitudinal Analysis and Mini-Review. PLoS ONE 2017, 12, e0170393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Johnson, R.J.; Bakris, G.L.; Borghi, C.; Chonchol, M.B.; Feldman, D.; Lanaspa, M.A.; Merriman, T.R.; Moe, O.W.; Mount, D.B.; Sanchez-Lozada, L.-G.; et al. Hyperuricemia, Acute and Chronic Kidney Disease, Hypertension, and Cardiovascular Disease: Report of a Scientific Workshop Organized by the National Kidney Foundation. Am. J. Kidney Dis. 2018, 71, 851–865. [Google Scholar] [CrossRef]
  7. Cho, S.K.; Chang, Y.; Kim, I.; Ryu, S. U-Shaped Association between Serum Uric Acid Level and Risk of Mortality: A Cohort Study. Arthritis Rheumatol. 2018, 70, 1122–1132. [Google Scholar] [CrossRef] [Green Version]
  8. Kim, S.Y.; Guevara, J.; Kim, K.M.; Choi, H.K.; Heitjan, D.F.; Albert, D.A. Hyperuricemia and coronary heart disease: A systematic review and meta-analysis. Arthritis Care Res. 2010, 62, 170–180. [Google Scholar] [CrossRef] [Green Version]
  9. Choi, H.K.; Curhan, G. Independent Impact of Gout on Mortality and Risk for Coronary Heart Disease. Circulation 2007, 116, 894–900. [Google Scholar] [CrossRef]
  10. Johnson, R.J.; Nakagawa, T.; Sanchez-Lozada, L.G.; Shafiu, M.; Sundaram, S.; Le, M.; Ishimoto, T.; Sautin, Y.Y.; Lanaspa, M.A. Sugar, Uric Acid, and the Etiology of Diabetes and Obesity. Diabetes 2013, 62, 3307–3315. [Google Scholar] [CrossRef] [Green Version]
  11. Li, L.; Yang, C.; Zhao, Y.; Zeng, X.; Liu, F.; Fu, P. Is hyperuricemia an independent risk factor for new-onset chronic kidney disease?: A systematic review and meta-analysis based on observational cohort studies. BMC Nephrol. 2014, 15, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Gupta, A.; Moore, J.A. Tumor Lysis Syndrome. JAMA Oncol. 2018, 4, 895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Li, C.; Hsieh, M.-C.; Chang, S.-J. Metabolic syndrome, diabetes, and hyperuricemia. Curr. Opin. Rheumatol. 2013, 25, 210–216. [Google Scholar] [CrossRef] [PubMed]
  14. Dehlin, M.; Jacobsson, L.; Roddy, E. Global epidemiology of gout: Prevalence, incidence, treatment patterns and risk factors. Nat. Rev. Rheumatol. 2020, 16, 380–390. [Google Scholar] [CrossRef]
  15. Cronstein, B.N.; Terkeltaub, R. The inflammatory process of gout and its treatment. Arthritis Res. Ther. 2006, 8, S3. [Google Scholar] [CrossRef] [Green Version]
  16. Dalbeth, N.; Clark, B.; Gregory, K.; Gamble, G.; Sheehan, T.; Doyle, A.; McQueen, F.M. Mechanisms of bone erosion in gout: A quantitative analysis using plain radiography and computed tomography. Ann. Rheum. Dis. 2009, 68, 1290–1295. [Google Scholar] [CrossRef]
  17. 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. 2011, 63, 3136–3141. [Google Scholar] [CrossRef]
  18. 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. 2019, 71, 991–999. [Google Scholar] [CrossRef]
  19. Kuo, C.-F.; Grainge, M.J.; Zhang, W.; Doherty, M. Global epidemiology of gout: Prevalence, incidence and risk factors. Nat. Rev. Rheumatol. 2015, 11, 649–662. [Google Scholar] [CrossRef]
  20. FitzGerald, J.D.; Dalbeth, N.; Mikuls, T.; Brignardello-Petersen, R.; Guyatt, G.; Abeles, A.M.; Gelber, A.C.; Harrold, L.R.; Khanna, D.; King, C.; et al. American College of Rheumatology Guideline for the Management of Gout. Arthritis Care Res. 2020, 72, 744–760. [Google Scholar] [CrossRef]
  21. Richette, P.; Doherty, M.; Pascual, E.; Barskova, V.; Becce, F.; Castañeda-Sanabria, J.; Coyfish, M.; Guillo, S.; Jansen, T.L.; Janssens, H.; et al. 2016 updated EULAR evidence-based recommendations for the management of gout. Ann. Rheum. Dis. 2017, 76, 29–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Graham, G.G.; Stocker, S.L.; Kannangara, D.R.; Day, R.O. Predicting Response or Non-response to Urate-Lowering Therapy in Patients with Gout. Curr. Rheumatol. Rep. 2018, 20, 47. [Google Scholar] [CrossRef] [PubMed]
  23. Cicero, A.F.G.; Fogacci, F.; Cincione, R.I.; Tocci, G.; Borghi, C. Clinical Effects of Xanthine Oxidase Inhibitors in Hyperuricemic Patients. Med. Princ. Pract. 2021, 30, 122–130. [Google Scholar] [CrossRef] [PubMed]
  24. PubChem Compound Summary for CID 5288320, Topiroxostat. National Center for Biotechnology Information. 2021. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Topiroxostat (accessed on 15 November 2021).
  25. White, W.B.; Saag, K.G.; Becker, M.A.; Borer, J.S.; Gorelick, P.B.; Whelton, A.; Hunt, B.; Castillo, M.; Gunawardhana, L.; CARES Investigators. Cardiovascular Safety of Febuxostat or Allopurinol in Patients with Gout. N. Engl. J. Med. 2018, 378, 1200–1210. [Google Scholar] [CrossRef]
  26. Pawar, A.; Desai, R.J.; Liu, J.; Kim, E.; Kim, S.C. Updated Assessment of Cardiovascular Risk in Older Patients with Gout Initiating Febuxostat Versus Allopurinol. J. Am. Heart Assoc. 2021, 10, e020045. [Google Scholar] [CrossRef]
  27. Mackenzie, I.S.; Ford, I.; Nuki, G.; Hallas, J.; Hawkey, C.J.; Webster, J.; Ralston, S.H.; Walters, M.; Robertson, M.; De Caterina, R.; et al. Long-term cardiovascular safety of febuxostat compared with allopurinol in patients with gout (FAST): A multicentre, prospective, randomised, open-label, non-inferiority trial. Lancet 2020, 396, 1745–1757. [Google Scholar] [CrossRef]
  28. Shannon, J.A.; Cole, S.W. Pegloticase: A Novel Agent for Treatment-Refractory Gout. Ann. Pharmacother. 2012, 46, 368–376. [Google Scholar] [CrossRef]
  29. Fleischmann, R.; Kerr, B.; Yeh, L.-T.; Suster, M.; Shen, Z.; Polvent, E.; Hingorani, V.; Quart, B.; Manhard, K.; Miner, J.N.; et al. Pharmacodynamic, pharmacokinetic and tolerability evaluation of concomitant administration of lesinurad and febuxostat in gout patients with hyperuricaemia. Rheumatology 2014, 53, 2167–2174. [Google Scholar] [CrossRef] [Green Version]
  30. Xu, S.; Liu, X.; Ming, J.; Chen, S.; Wang, Y.; Liu, X.; Liu, H.; Peng, Y.; Wang, J.; Lin, J.; et al. A phase 3, multicenter, randomized, allopurinol-controlled study assessing the safety and efficacy of oral febuxostat in Chinese gout patients with hyperuricemia. Int. J. Rheum. Dis. 2015, 18, 669–678. [Google Scholar] [CrossRef]
  31. Becker, M.A.; Schumacher, H.R., Jr.; Wortmann, R.L.; Macdonald, P.A.; Eustace, D.; Palo, W.A.; Streit, J.; Joseph-Ridge, N. Febuxostat Compared with Allopurinol in Patients with Hyperuricemia and Gout. N. Engl. J. Med. 2005, 353, 2450–2461. [Google Scholar] [CrossRef] [Green Version]
  32. Schumacher, H.R., Jr.; Becker, M.A.; Wortmann, R.L.; Macdonald, P.A.; Hunt, B.; Streit, J.; Lademacher, C.; Joseph-Ridge, N. 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 Rheum. 2008, 59, 1540–1548. [Google Scholar] [CrossRef] [PubMed]
  33. Becker, M.A.; Schumacher, H.R.; Espinoza, L.R.; Wells, A.F.; MacDonald, P.; Lloyd, E.; Lademacher, C. The urate-lowering efficacy and safety of febuxostat in the treatment of the hyperuricemia of gout: The CONFIRMS trial. Arthritis Res. Ther. 2010, 12, R63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Becker, M.A.; Fitz-Patrick, D.; Choi, H.K.; Dalbeth, N.; Storgard, C.; Cravets, M.; Baumgartner, S. An open-label, 6-month study of allopurinol safety in gout: The LASSO study. Semin. Arthritis Rheum. 2015, 45, 174–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Huang, X.; Du, H.; Gu, J.; Zhao, D.; Jiang, L.; Li, X.; Zuo, X.; Liu, Y.; Li, Z.; Li, X.; et al. An allopurinol-controlled, multicenter, randomized, double-blind, parallel between-group, comparative study of febuxostat in Chinese patients with gout and hyperuricemia. Int. J. Rheum. Dis. 2014, 17, 679–686. [Google Scholar] [CrossRef] [PubMed]
  36. 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, 290–297. [Google Scholar] [CrossRef] [PubMed]
  37. 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. Rheumatol. 2017, 36, 649–656. [Google Scholar] [CrossRef] [Green Version]
  38. Tausche, A.-K.; Christoph, M.; Forkmann, M.; Richter, U.; Kopprasch, S.; Bielitz, C.; Aringer, M.; Wunderlich, C. As compared to allopurinol, urate-lowering therapy with febuxostat has superior effects on oxidative stress and pulse wave velocity in patients with severe chronic tophaceous gout. Rheumatol. Int. 2014, 34, 101–109. [Google Scholar] [CrossRef]
  39. Shelmadine, B.; Bowden, R.G.; Wilson, R.L.; Beavers, D.; Hartman, J. The effects of lowering uric acid levels using allopurinol on markers of metabolic syndrome in end-stage renal disease patients: A pilot study. Anadolu Kardiyol. Derg. 2009, 9, 385–389. [Google Scholar]
  40. Perez-Ruiz, F.; Alonso-Ruiz, A.; Calabozo, M.; Herrero-Beites, A.; Garcia-Erauskin, G.; Ruiz-Lucea, E. Efficacy of allopurinol and benzbromarone for the control of hyperuricaemia. A pathogenic approach to the treatment of primary chronic gout. Ann. Rheum. Dis. 1998, 57, 545–549. [Google Scholar] [CrossRef]
  41. Omrani, H.; Muhammad, S.; Sadeghi, K.; Raeisi, D. The effect of allopurinol treatment regimen on serum uric acid and arterial blood pressure in hemodialysis patients. J. Kermanshah Univ. Med. Sci. 2016, 20, 56–61. [Google Scholar]
  42. Jalalzadeh, M.; Nurcheshmeh, Z.; Mohammadi, R.; Mousavinasab, N.; Ghadiani, M.H. The effect of allopurinol on lowering blood pressure in hemodialysis patients with hyperuricemia. J. Res. Med. Sci. 2012, 17, 1039–1046. [Google Scholar] [PubMed]
  43. Golmohammadi, S.; Almasi, A.; Manouchehri, M.; Omrani, H.R.; Zandkarimi, M.R. Allopurinol Against Progression of Chronic Kidney Disease. Iran. J. Kidney Dis. 2017, 11, 286–293. [Google Scholar] [PubMed]
  44. Goicoechea, M.; De Vinuesa, S.G.; Verdalles, U.; Ruiz-Caro, C.; Ampuero, J.; Rincón, A.; Arroyo, D.; Luño, J. Effect of Allopurinol in Chronic Kidney Disease Progression and Cardiovascular Risk. Clin. J. Am. Soc. Nephrol. 2010, 5, 1388–1393. [Google Scholar] [CrossRef]
  45. Baumgartner, S.; Yeh, L.-T.; Shen, Z.; Kerr, B.; Manhard, K.; Quart, B. The Effect of Lesinurad in Combination with Allopurinol on Serum Uric Acid Levels in Patients with Gout. J. Clin. Pharmacol. 2018, 58, 1164–1170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Reinders, M.K.; Haagsma, C.; Jansen, T.L.; Van Roon, E.N.; Delsing, J.; Van De Laar, M.A.F.J.; Brouwers, J.R.B.J. A randomised controlled trial on the efficacy and tolerability with dose escalation of allopurinol 300–600 mg/day versus benzbromarone 100–200 mg/day in patients with gout. Ann. Rheum. Dis. 2009, 68, 892–897. [Google Scholar] [CrossRef]
  47. Reinders, M.; Van Roon, E.; Jansen, T.L.; Delsing, J.; Griep, E.; Hoekstra, M.; Van der Laar, M.A.F.J.; Brouwers, J.R.B.J. Efficacy and tolerability of urate-lowering drugs in gout: A randomised controlled trial of benzbromarone versus probenecid after failure of allopurinol. Ann. Rheum. Dis. 2009, 68, 51–56. [Google Scholar] [CrossRef]
  48. Siu, Y.-P.; Leung, K.-T.; Tong, M.K.H.; Kwan, T.-H. Use of Allopurinol in Slowing the Progression of Renal Disease through Its Ability to Lower Serum Uric Acid Level. Am. J. Kidney Dis. 2006, 47, 51–59. [Google Scholar] [CrossRef] [Green Version]
  49. Yelken, B.; Caliskan, Y.; Gorgulu, N.; Altun, I.; Yilmaz, A.; Yazici, H.; Oflaz, H.; Yildiz, A. Reduction of uric acid levels with allopurinol treatment improves endothelial function in patients with chronic kidney disease. Clin. Nephrol. 2012, 77, 275–282. [Google Scholar] [CrossRef]
  50. Nagaoka, Y.; Tanaka, Y.; Yoshimoto, H.; Suzuki, R.; Ryu, K.; Ueda, M.; Akiyama, M.; Nagai, M.; Miyaoka, Y.; Kanda, E.; et al. The effect of small dose of topiroxostat on serum uric acid in patients receiving hemodialysis. Hemodial. Int. 2018, 22, 388–393. [Google Scholar] [CrossRef]
  51. Becker, M.A.; Schumacher, H.R.; Wortmann, R.L.; Macdonald, P.A.; Palo, W.A.; Eustace, D.; Vernillet, L.; Joseph-Ridge, N. Febuxostat, a novel nonpurine selective inhibitor of xanthine oxidase: A twenty-eight-day, multicenter, phase II, randomized, double-blind, placebo-controlled, dose-response clinical trial examining safety and efficacy in patients with gout. Arthritis Rheum. 2005, 52, 916–923. [Google Scholar] [CrossRef]
  52. Schumacher, J.H.R.; Becker, M.A.; Lloyd, E.; MacDonald, P.A.; Lademacher, C. Febuxostat in the treatment of gout: 5-yr findings of the FOCUS efficacy and safety study. Rheumatology 2009, 48, 188–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kamatani, N.; Fujimori, S.; Hada, T.; Hosoya, T.; Kohri, K.; Nakamura, T.; Ueda, T.; Yamamoto, T.; Yamanaka, H.; Matsuzawa, Y. Placebo-controlled double-blind dose-response study of the non-purine-selective xanthine oxidase inhibitor febuxostat (TMX-67) in patients with hyperuricemia (including gout patients) in japan: Late phase 2 clinical study. J. Clin. Rheumatol. Pract. Rep. Rheum. Musculoskelet. Dis. 2011, 17 (Suppl. 2), S35–S43. [Google Scholar]
  54. Kamatani, N.; Fujimori, S.; Hada, T.; Hosoya, T.; Kohri, K.; Nakamura, T.; Ueda, T.; Yamamoto, T.; Yamanaka, H.; Matsuzawa, Y. An allopurinol-controlled, multicenter, randomized, open-label, parallel between-group, comparative study of febuxostat (TMX-67), a non-purine-selective inhibitor of xanthine oxidase, in patients with hyperuricemia including those with gout in Japan: Phase 2 exploratory clinical study. J. Clin. Rheumatol. Pract. Rep. Rheum. Musculoskelet. Dis. 2011, 17 (Suppl. 2), S44–S49. [Google Scholar]
  55. Kamatani, N.; Fujimori, S.; Hada, T.; Hosoya, T.; Kohri, K.; Nakamura, T.; Ueda, T.; Yamamoto, T.; Yamanaka, H.; Matsuzawa, Y. An allopurinol-controlled, randomized, double-dummy, double-blind, parallel between-group, comparative study of febuxostat (TMX-67), a non-purine-selective inhibitor of xanthine oxidase, in patients with hyperuricemia including those with gout in Japan: Phase 3 clinical study. J. Clin. Rheumatol. Pract. Rep. Rheum. Musculoskelet. Dis. 2011, 17 (Suppl. 2), S13–S18. [Google Scholar]
  56. Kamatani, N.; Fujimori, S.; Hada, T.; Hosoya, T.; Kohri, K.; Nakamura, T.; Ueda, T.; Yamamoto, T.; Yamanaka, H.; Matsuzawa, Y. Placebo-controlled, double-blind study of the non-purine-selective xanthine oxidase inhibitor Febuxostat (TMX-67) in patients with hyperuricemia including those with gout in Japan: Phase 3 clinical study. J. Clin. Rheumatol. Pract. Rep. Rheum. Musculoskelet. Dis. 2011, 17 (Suppl. 2), S19–S26. [Google Scholar]
  57. Hosoya, T.; Ohno, I. A repeated oral administration study of febuxostat (TMX-67), a non-purine-selective inhibitor of xanthine oxidase, in patients with impaired renal function in Japan: Pharmacokinetic and pharmacodynamic study. J. Clin. Rheumatol. Pract. Rep. Rheum. Musculoskelet. Dis. 2011, 17 (Suppl. 2), S27–S34. [Google Scholar]
  58. Tanaka, A.; Nakamura, T.; Sato, E.; Node, K. Clinical Effects of Topiroxostat on Renal and Endothelial Function in A Patient with Chronic Kidney Disease and Hyperuricemic Arteriolopathy: A Case Report. Drugs R D 2017, 17, 97–101. [Google Scholar] [CrossRef] [Green Version]
  59. Hosoya, T.; Ohno, I.; Nomura, S.; Hisatome, I.; Uchida, S.; Fujimori, S.; Yamamoto, T.; Hara, S. Effects of topiroxostat on the serum urate levels and urinary albumin excretion in hyperuricemic stage 3 chronic kidney disease patients with or without gout. Clin. Exp. Nephrol. 2014, 18, 876–884. [Google Scholar] [CrossRef] [Green Version]
  60. Hosoya, T.; Sasaki, T.; Hashimoto, H.; Sakamoto, R.; Ohashi, T. Clinical efficacy and safety of topiroxostat in Japanese male hyperuricemic patients with or without gout: An exploratory, phase 2a, multicentre, randomized, double-blind, placebo-controlled study. J. Clin. Pharm. Ther. 2016, 41, 298–305. [Google Scholar] [CrossRef]
  61. Wada, T.; Hosoya, T.; Honda, D.; Sakamoto, R.; Narita, K.; Sasaki, T.; Okui, D.; Kimura, K. Uric acid-lowering and renoprotective effects of topiroxostat, a selective xanthine oxidoreductase inhibitor, in patients with diabetic nephropathy and hyperuricemia: A randomized, double-blind, placebo-controlled, parallel-group study (UPWARD study). Clin. Exp. Nephrol. 2018, 22, 860–870. [Google Scholar] [CrossRef] [Green Version]
  62. Tausche, A.-K.; Alten, R.; Dalbeth, N.; Kopicko, J.; Fung, M.; Adler, S.; Bhakta, N.; Storgard, C.; Baumgartner, S.; Saag, K. Lesinurad monotherapy in gout patients intolerant to a xanthine oxidase inhibitor: A 6 month phase 3 clinical trial and extension study. Rheumatology 2017, 56, 2170–2178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gibson, T.; Simmonds, H.A.; Potter, C.; Rogers, V. A Controlled Study of the Effect of Long Term Allopurinol Treatment on Renal Function in Gout. Adv. Exp. Med. Biol. 1980, 122, 257–262. [Google Scholar]
  64. Singal, K.K.; Goyal, S.; Gupta, P.; Aggawal, B.K. Comparison between Allopurinol and Febuxostat in management of gout patients—A prospective study. Bangladesh J. Med. Sci. 2011, 10, 257–259. [Google Scholar] [CrossRef] [Green Version]
  65. Dalbeth, N.; Kumar, S.; Stamp, L.; Gow, P. Dose adjustment of allopurinol according to creatinine clearance does not provide adequate control of hyperuricemia in patients with gout. J. Rheumatol. 2006, 33, 1646–1650. [Google Scholar] [PubMed]
  66. Stamp, L.K.; Chapman, P.T.; Barclay, M.; Horne, A.; Frampton, C.; Tan, P.; Drake, J.; Dalbeth, N. The effect of kidney function on the urate lowering effect and safety of increasing allopurinol above doses based on creatinine clearance: A post hoc analysis of a randomized controlled trial. Arthritis Res. Ther. 2017, 19, 283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Stamp, L.K.; Chapman, P.T.; Barclay, M.; Horne, A.; Frampton, C.; Tan, P.; Drake, J.; Dalbeth, N. 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. 2017, 76, 1522–1528. [Google Scholar] [CrossRef]
  68. Ye, P.; Yang, S.; Zhang, W.; Lv, Q.; Cheng, Q.; Mei, M.; Luo, T.; Liu, L.; Chen, S.; Li, Q. Efficacy and Tolerability of Febuxostat in Hyperuricemic Patients with or without Gout: A Systematic Review and Meta-Analysis. Clin. Ther. 2013, 35, 180–189. [Google Scholar] [CrossRef]
  69. Borghi, C.; Perez-Ruiz, F. Urate lowering therapies in the treatment of gout: A systematic review and meta-analysis. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 983–992. [Google Scholar]
  70. Li, S.; Yang, H.; Guo, Y.; Wei, F.; Yang, X.; Li, D.; Li, M.; Xu, W.; Li, W.; Sun, L.; et al. Comparative efficacy and safety of urate-lowering therapy for the treatment of hyperuricemia: A systematic review and network meta-analysis. Sci. Rep. 2016, 6, 33082. [Google Scholar] [CrossRef] [Green Version]
  71. Gandhi, P.K.; Gentry, W.M.; Ma, Q.; Bottorff, M.B. Cost-Effectiveness Analysis of Allopurinol versus Febuxostat in Chronic Gout Patients: A U.S. Payer Perspective. J. Manag. Care Spec. Pharm. 2015, 21, 165–175. [Google Scholar] [CrossRef] [Green Version]
  72. Jutkowitz, E.; Choi, H.K.; Pizzi, L.T.; Kuntz, K.M. Cost-Effectiveness of Allopurinol and Febuxostat for the Management of Gout. Ann. Intern. Med. 2014, 161, 617–626. [Google Scholar] [CrossRef] [PubMed]
  73. Takeda Pharmaceuticals America, Inc. Uloric (Febuxostat) [Package Insert]; Takeda Pharmaceuticals America, Inc.: Chicago, IL, USA, 2009. [Google Scholar]
  74. Tassaneeyakul, W.; Jantararoungtong, T.; Chen, P.; Lin, P.-Y.; Tiamkao, S.; Khunarkornsiri, U.; Chucherd, P.; Konyoung, P.; Vannaprasaht, S.; Choonhakarn, C.; et al. Strong association between HLA-B*5801 and allopurinol-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in a Thai population. Pharmacogenet. Genom. 2009, 19, 704–709. [Google Scholar] [CrossRef] [PubMed]
  75. Hung, S.-I.; Chung, W.-H.; Liou, L.-B.; Chu, C.-C.; Lin, M.; Huang, H.-P.; Lin, Y.-L.; Lan, J.-L.; Yang, L.-C.; Hong, H.-S.; et al. HLA-B*5801 allele as a genetic marker for severe cutaneous adverse reactions caused by allopurinol. Proc. Natl. Acad. Sci. USA 2005, 102, 4134–4139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Roberts, R.L.; Wallace, M.C.; Phipps-Green, A.; Topless, R.; Drake, J.M.; Tan, P.; Dalbeth, N.; Merriman, T.; Stamp, L.K. ABCG2 loss-of-function polymorphism predicts poor response to allopurinol in patients with gout. Pharmacogenomics J. 2017, 17, 201–203. [Google Scholar] [CrossRef] [PubMed]
  77. Sarawate, C.A.; Brewer, K.K.; Yang, W.; Patel, P.A.; Schumacher, H.R.; Saag, K.G.; Bakst, A.W. Gout medication treatment patterns and adherence to standards of care from a managed care perspective. Mayo Clin. Proc. 2006, 81, 925–934. [Google Scholar] [CrossRef]
  78. Cho, S.K.; Winkler, C.A.; Lee, S.-J.; Chang, Y.; Ryu, S. The Prevalence of Hyperuricemia Sharply Increases from the Late Menopausal Transition Stage in Middle-Aged Women. J. Clin. Med. 2019, 8, 296. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Flow diagram. Summary of literature search and selection process. Note. CKD: chronic kidney disease; QD: quarter in die (four times a day); SUA: serum uric acid; and ULT: uric lowering therapy. a One study was listed as a reference for an article sourced through Embase [27].
Figure 1. Flow diagram. Summary of literature search and selection process. Note. CKD: chronic kidney disease; QD: quarter in die (four times a day); SUA: serum uric acid; and ULT: uric lowering therapy. a One study was listed as a reference for an article sourced through Embase [27].
Jcm 11 02468 g001
Figure 2. Forest Plot of Achievement Difference in Febuxostat Monotherapy vs. Allopurinol 300 mg QD–SUA < 6 mg/dL [30,31,35,64].
Figure 2. Forest Plot of Achievement Difference in Febuxostat Monotherapy vs. Allopurinol 300 mg QD–SUA < 6 mg/dL [30,31,35,64].
Jcm 11 02468 g002
Table 1. Meta-analysis of Serum uric acid-lowering effect of approved uric lowering therapy drugs with daily doses [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].
Table 1. Meta-analysis of Serum uric acid-lowering effect of approved uric lowering therapy drugs with daily doses [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].
ULT Drugs
AllopurinolFebuxostatTopiroxostat
βSEpβSEpβSEp
n29 28 16
τ226.35 24.03 17.25
I2 residual (%)91.93 85.09 89.03
Adjusted R20.60 0.70 0.67
Intercept17.522.03<0.00131.972.11<0.00123.311.88<0.001
Dose0.050.01<0.0010.180.03<0.0010.100.02<0.001
Dose Range50–43520–24020–152
Legend. Shown are the beta values for sample size and R2 (the proportion of the variation in the dependent variable that is explained by the independent variable), residual, and R2 adjusted for chronic kidney disease status, sample size, length of follow-up, data collection site, and proportion of male participants. For the Y intercept and dose, beta coefficient, and standard errors of the intercept are also shown. Also shown are the range of typical daily doses for each drug.
Table 2. Covariate model for allopurinol and post-hoc analyses based on data collection site [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].
Table 2. Covariate model for allopurinol and post-hoc analyses based on data collection site [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].
Reference Collection SiteMiddle EastMulti-NationalNorth America
βSEpβSEpβSEp
Intercept−46.3620.180.051−109.2032.540.010−102.3932.470.014
Dose0.360.070.0010.360.070.0010.360.070.001
Chronic kidney
disease (CKD)
diagnosis
27.6212.710.06227.6212.710.06227.6212.710.062
Sample size0.0020.010.7710.0020.010.7710.0020.010.771
Follow-up (days)0.030.020.0900.030.020.0900.030.020.090
Region
China−26.394.910.00136.4511.980.01629.6510.990.027
Europe−109.9823.920.002−47.1414.280.011−53.9511.840.002
Japan15.915.610.02278.7518.550.00371.9517.570.003
Middle Eastern---62.8414.100.00256.0312.830.002
Multi-National−62.8414.100.002----6.816.210.305
North America−56.0312.830.0026.846.210.305---
Proportion of males−8.426.490.231−8.426.490.231−8.426.490.231
Legend. The covariate model used each clinical trial location as a reference group: n = 19, τ2 = 7.32, I2 residual (%) = 76.74, adjusted R2 = 0.91. The dose-dependent serum uric acid-lowering effect was estimated while accounting for CKD status, sample size, length of follow-up, data collection site, and proportion of male participants. CKD was defined as eGFR < 60 mL/min/1.73 m2, CrCl < 50 mL/min/1.73 m2, or serum creatinine > 1.5 mg/dL. The proportion of male participants was categorized by the median score for all arms: 0 for less male-dominant arms and 1 for more male-dominant arms. Region refers to the site where clinical trials were conducted. Follow-up refers to the duration of the clinical trial.
Table 3. Covariate model for febuxostat and post-hoc analyses based on data collection site [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].
Table 3. Covariate model for febuxostat and post-hoc analyses based on data collection site [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].
Reference Collection SiteChinaEuropeJapanNorth America
βSEpβSEpβSEpβSEp
Intercept18.856.940.01523.237.740.00830.683.90<0.00128.144.29<0.001
Dose0.230.04<0.0010.230.04<0.0010.230.04<0.0010.230.04<0.001
Chronic kidney
disease (CKD)
diagnosis
−9.679.050.301−9.679.050.301−9.679.050.301−9.679.050.301
Sample size0.0030.010.8010.0030.010.8010.0030.010.8010.0030.010.801
Duration (days)0.020.030.4920.020.030.4920.020.030.4920.020.030.492
Region
China---−4.389.710.658−11.835.020.032−9.295.820.130
Europe4.389.710.658---−7.458.610.400−4.917.840.540
Japan11.835.020.0327.458.610.400---2.543.610.492
North America9.295.820.1304.917.840.540−2.543.610.492---
Male proportion0.263.070.9330.263.070.9330.263.070.9330.263.070.933
Legend: The covariate model used each clinical trial location as a reference group: n = 25, τ2 = 15.55, I2 residual (%) = 71.84, adjusted R2 = 0.81. The dose-dependent serum uric acid-lowering effect was estimated while accounting for CKD status, sample size, length of follow-up, data collection site, and proportion of male participants. CKD was defined as eGFR < 60 mL/min/1.73 m2, creatinine clearance < 50 mL/min/1.73 m2 [correct?), or serum creatinine > 1.5 mg/dL. The proportion of male participants was categorized by the median score for all arms: 0 for less male-dominant arms and 1 for more male-dominant arms. Region refers to the site where clinical trials were conducted. Duration refers to the length of the clinical trial.
Table 4. Covariate model for topiroxostat and post-hoc analyses based on proportion of male participants [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].
Table 4. Covariate model for topiroxostat and post-hoc analyses based on proportion of male participants [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].
JapanMore Male-DominantLess Male-Dominant
βSEpβSEpβSEp
n16 8 8
τ210.57 1.71 2.81
I2 residual (%)82.55 34.25 70.03
Adjusted R280.06 97.07 94.73
Intercept30.2310.570.017−34.5315.730.11721.737.180.039
Dose0.120.030.0030.110.040.0580.030.060.581
Chronic
kidney
disease (CKD)
diagnosis
−10.935.700.084−23.134.350.013
Sample size−0.0080.060.902−0.080.040.1720.280.060.231
Follow-up (days)1.092.800.70513.914.450.0520.111.800.954
Male proportion−14.225.160.020
Legend: The dose-dependent serum uric acid-lowering effect was estimated while accounting for CKD status, sample size, length of follow-up, data collection site, and proportion of male participants. CKD was defined as eGFR < 60 mL/min/1.73 m2, CrCl < 50 mL/min/1.73 m2, or serum creatinine > 1.5 mg/dL. The proportion of male participants was categorized by the median score for all arms: 0 for less male-dominant arms and 1 for more male-dominant arms. Region refers to the site where clinical trials were conducted. Follow-up refers to the length of the clinical trial.
Table 5. Serum uric acid-lowering Achievement Difference vs. allopurinol 300 mg daily.
Table 5. Serum uric acid-lowering Achievement Difference vs. allopurinol 300 mg daily.
Study
(Year; NCT)
Achieved Outcome GoalDrug/Dose (mg)AD vs. Allopurinol 300 mg Daily (95% CI)AD vs. Allopurinol 300 mg Daily by Dose (95% CI)
Xu et al.
(2015; NCT02082769) [30]
SUA < 6 mg/dL at last three visits
(6 months of treatment) **
Febuxostat 40 mg daily0.06 (−0.032, 0.142)0.05 (−0.018, 0.109)
Huang et al. (2014) [35]SUA < 6 mg/dL at last three monthly visits
(7 months of treatment)
Febuxostat 40 mg daily0.04 (−0.057, 0.127)
Xu et al.
(2015; NCT02082769) [30]
SUA < 6 mg/dL at last three visits
(6 months of treatment) **
Febuxostat 80 mg daily0.17 (0.072, 0.260) *0.25 (0.165, 0.325) *
Singal et al. (2011) [64]SUA < 6 mg/dL at last three measurements
(6 months of treatment)
Febuxostat 80 mg daily0.30 (0.118, 0.482) *
Huang et al. (2014) [35]SUA < 6 mg/dL at last three monthly visits
(7 months of treatment)
Febuxostat 80 mg daily0.21 (0.111, 0.307) *
Becker et al.
(2005; NCT00102440) [31]
SUA < 6 mg/dL at last three monthly visits
(1 year of treatment)
Febuxostat 80 mg daily0.32 (0.243, 0.402) *
Becker et al.
(2005; NCT00102440) [31]
SUA < 6 mg/dL at last three monthly visits
(1 year of treatment)
Febuxostat 120 mg daily0.40 (0.326, 0.483) *0.40 (0.326, 0.483) *
Legend: * statistically significant; ** converted from 356.9 µmol/L using 59.48 µmol/L = 1 mg/dL. NCT: the national clinical trial number.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lee, Y.; Hwang, J.; Desai, S.H.; Li, X.; Jenkins, C.; Kopp, J.B.; Winkler, C.A.; Cho, S.K. Efficacy of Xanthine Oxidase Inhibitors in Lowering Serum Uric Acid in Chronic Kidney Disease: A Systematic Review and Meta-Analysis. J. Clin. Med. 2022, 11, 2468. https://doi.org/10.3390/jcm11092468

AMA Style

Lee Y, Hwang J, Desai SH, Li X, Jenkins C, Kopp JB, Winkler CA, Cho SK. Efficacy of Xanthine Oxidase Inhibitors in Lowering Serum Uric Acid in Chronic Kidney Disease: A Systematic Review and Meta-Analysis. Journal of Clinical Medicine. 2022; 11(9):2468. https://doi.org/10.3390/jcm11092468

Chicago/Turabian Style

Lee, Yoojin, Jennifer Hwang, Shaan H. Desai, Xiaobai Li, Christopher Jenkins, Jeffrey B. Kopp, Cheryl A. Winkler, and Sung Kweon Cho. 2022. "Efficacy of Xanthine Oxidase Inhibitors in Lowering Serum Uric Acid in Chronic Kidney Disease: A Systematic Review and Meta-Analysis" Journal of Clinical Medicine 11, no. 9: 2468. https://doi.org/10.3390/jcm11092468

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop