Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Pharmacol., 01 October 2025

Sec. Inflammation Pharmacology

Volume 16 - 2025 | https://doi.org/10.3389/fphar.2025.1660890

Advances in drug delivery systems for the management of gout and hyperuricemia

Zining Peng,,&#x;Zining Peng1,2,3Fanyu Meng&#x;Fanyu Meng1Qian Deng&#x;Qian Deng1Yuanbo Huang,Yuanbo Huang1,3Danning MaoDanning Mao1Yuan LongYuan Long1Weitian YanWeitian Yan2Jiangyun Peng
Jiangyun Peng2*Xingqiang Wang,
Xingqiang Wang2,4*Nian Liu,,
Nian Liu1,2,4*
  • 1The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
  • 2Department of Rheumatology, The No.1 Affiliated Hospital of Yunnan University of Chinese Medicine/Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
  • 3Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming, China
  • 4Yunnan Provincial Clinical Medicine Research Center of Rheumatism in TCM, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China

Gout and hyperuricemia represent significant global health burdens, characterized by painful inflammatory arthritis and systemic metabolic dysfunction, respectively. Current pharmacological management faces substantial limitations, including poor bioavailability, systemic toxicity, narrow therapeutic indices, immunogenicity, and suboptimal patient adherence due to frequent dosing and adverse effects. These challenges underscore the critical need for innovative therapeutic strategies. Advanced drug delivery systems (DDSs) have emerged as transformative solutions to overcome these hurdles. This comprehensive review critically analyzes recent advances in DDSs tailored to the management of gout and hyperuricemia. We first elucidate the intricate pathophysiological mechanisms linking hyperuricemia, monosodium urate (MSU) crystal deposition, NLRP3 inflammasome activation, and chronic inflammation. We then systematically explore cutting-edge DDS platforms, including lipid-based, polymer-based, and other systems. These engineered drug delivery systems significantly enhance therapeutic outcomes in gout and hyperuricemia by improving drug solubility, enabling targeted delivery, providing sustained release, facilitating synergistic drug co-delivery, and responding to pathological microenvironments, although preclinical evidence is limited and clinical evidence supporting their efficacy and safety remains sparse. Finally, we highlight translational challenges and future directions while emphasizing the considerable promise of integrating AI, biomaterial science, and personalized medicine to advance patient-centric DDS. Although progress has been made, sustained interdisciplinary collaboration and rigorous clinical validation remain critical to translate these innovations into tangible improvements in long-term disease management and quality of life for patients with gout and hyperuricemia.

1 Introduction

Hyperuricemia, defined as elevated serum urate (SU) levels (>6.8 mg/dL), is a metabolic disorder affecting approximately 20% of the global population, with its prevalence increasing due to dietary changes, obesity, and aging populations (Nakayama et al., 2023; Zhong et al., 2024). Gout, the most common inflammatory arthritis worldwide, arises from the deposition of monosodium urate (MSU) crystals in joints and soft tissues, driven by chronic hyperuricemia (Cabau et al., 2023; Nakayama et al., 2023). Epidemiological studies highlight a strong association between hyperuricemia/gout and comorbidities such as cardiovascular diseases, chronic kidney disease, and metabolic syndrome, further amplifying their clinical and socioeconomic burden (Cabau et al., 2023; Nakayama et al., 2023). Genetic predispositions, including mutations in urate transporters (e.g., URAT1), also contribute to disease susceptibility (Sun et al., 2024). Alarmingly, the global incidence of gout has increased significantly in recent decades, particularly in the Western and Asia–Pacific regions, underscoring the need for improved management strategies (Nakayama et al., 2023; Zhong et al., 2024; Dalbeth et al., 2021).

Current clinical guidelines for hyperuricemia and gout emphasize a dual approach: (1) acute flare management using nonsteroidal anti-inflammatory drugs (NSAIDs), colchicine, or corticosteroids and (2) long-term urate-lowering therapy (ULT) with xanthine oxidase inhibitors (e.g., allopurinol and febuxostat) or uricosurics (e.g., probenecid) (Karantas et al., 2024; Sattui and Gaffo, 2016). Despite these options, significant limitations persist. Allopurinol, the first-line ULT, suffers from reduced efficacy in patients with impaired conversion to its active metabolite, oxypurinol (OXY), and carries risks of severe hypersensitivity reactions (Mohamad et al., 2025; Yi et al., 2024). Oral and injectable formulations of existing drugs often exhibit poor bioavailability, systemic toxicity, and suboptimal patient adherence due to frequent dosing and adverse effects (e.g., gastrointestinal distress and rashes) (Sattui and Gaffo, 2016). For instance, the narrow therapeutic index of colchicine necessitates cautious dosing to prevent toxicity, while uricase therapies (e.g., pegloticase) are challenged by immunogenicity and instability (Cabau et al., 2023; Yang Y. et al., 2023).

Moreover, traditional drug delivery systems (DDSs)—primarily oral tablets and intravenous injections—fail to address localized MSU crystal deposition or provide sustained drug release, leading to recurrent flares and chronic joint damage (Mohamad et al., 2025; Yi et al., 2024). The lack of targeted delivery exacerbates off-target effects and limits therapeutic efficacy, particularly in patients with comorbidities requiring polypharmacy (Karantas et al., 2024). These shortcomings highlight the critical need for innovative DDSs to enhance drug safety, bioavailability, and patient compliance (Ezike et al., 2023).

Advanced DDSs offer transformative solutions to overcome the limitations of conventional therapies. By leveraging nanotechnology, biomaterials, and controlled-release mechanisms, modern DDSs provide the following therapeutic advantages. First, enhanced bioavailability: nanocarriers (e.g., liposomes and polymeric nanoparticles) improve solubility and stability of poorly water-soluble drugs like OXY and colchicine, ensuring consistent therapeutic plasma levels (Peng et al., 2023; Singh et al., 2017). Second, targeted delivery: ligand-functionalized carriers selectively accumulate drugs in inflamed joints or renal tissues, minimizing systemic exposure and adverse effects (Ezike et al., 2023; Yang B. et al., 2023). For example, transdermal microneedles loaded with allopurinol bypass hepatic metabolism, reducing first-pass effects and enabling sustained release (Chen et al., 2021). Third, sustained and controlled release: hydrogels and 3D-printed porous scaffolds (e.g., tantalum-based systems) provide localized, long-term drug delivery, ideal for managing chronic gout and preventing crystal recurrence (Hua et al., 2021; Wang et al., 2025). Fourth, combination therapy: co-delivery systems (e.g., lipid-based vesicles co-encapsulating anti-inflammatory and urate-lowering agents) synergistically address inflammation and hyperuricemia, simplifying treatment regimens (Li et al., 2023).

Recent advancements in DDSs have yielded promising preclinical and clinical outcomes (Jafernik et al., 2023). Transdermal and topical systems: ethosomes and transfersomes loaded with capsaicin (CAP) and triamcinolone acetonide (TCS) enhance skin permeation, offering localized analgesia and anti-inflammatory effects in acute gout (Singh et al., 2017). Similarly, dissolving microneedles (DMNs) for allopurinol delivery demonstrate sustained SU reduction in animal models, avoiding gastrointestinal toxicity (Singh et al., 2017). Nanoformulations: nonionic surfactant-based niosomes encapsulating allopurinol achieved 82% drug release within 24 h and significantly reduced SU levels in MSU crystal-induced gout models, outperforming free drug formulations (Singh et al., 2017). Lipid–polymer hybrid nanoparticles and inorganic carriers (e.g., silica nanoparticles) also improve uricase stability and targeting (Wang et al., 2025). Smart and responsive systems: pH-sensitive hydrogels and enzyme-responsive nanocapsules release drugs selectively in acidic or inflamed microenvironments, enhancing precision (Wang et al., 2025; Liu et al., 2024). For instance, a colchicine-coordinated nanogel system reduced the dosing frequency and mitigated systemic toxicity in chronic gout models (Yang B. et al., 2023). Biological and biomimetic systems: erythrocyte membrane-coated nanoparticles and macrophage-derived exosomes exploit natural trafficking mechanisms to deliver drugs to synovial tissues, demonstrating reduced immunogenicity (Wang et al., 2025).

This review presents a synthesized perspective on the pathophysiological mechanisms and therapeutic strategies for gout and hyperuricemia, with a nuanced focus on delineating the evolving role of DDS. While acknowledging the current reliance on preclinical studies and limited clinical validation data, this work underscores the promising potential of DDS-driven approaches to advance precision medicine frameworks. By integrating cutting-edge nanomedicine approaches, we critically analyze the molecular targets underpinning urate crystal deposition, NLRP3 inflammasome activation, and chronic inflammatory cascades in gout while contextualizing hyperuricemia as a modifiable risk factor. The manuscript highlights how DDSs—including stimuli-responsive nanoparticles, targeted ligands, and biodegradable carriers—enhance solubility, bioavailability, and tissue-specific delivery of urate-lowering agents (e.g., allopurinol and febuxostat) and anti-inflammatory therapies (e.g., colchicine and IL-1β inhibitors) (Chen Z. et al., 2025; Liu S. et al., 2023). By integrating mechanistic discoveries with emerging therapeutic innovations, the analysis highlights how DDS research is laying critical groundwork for future translational efforts. Although widespread clinical implementation awaits further validation through large-scale trials, these findings offer valuable insights to guide early-stage translational research and inform evidence-based decision-making in the evolving landscape of gout and hyperuricemia management.

2 Pathogenesis and pathophysiology of gout and hyperuricemia

2.1 Pathogenesis and pathophysiology of gout

Gout, a prototypical disorder of metabolic–immune dysregulation, is an inflammatory arthritis triggered by the deposition of MSU crystals. Its pathophysiology centers on chronic hyperuricemia and dysregulated uric acid metabolism (Bhadouria et al., 2025; Stiburkova and Ichida, 2025; Zhao et al., 2022). This section elaborates on its pathogenesis from multiple perspectives, including molecular mechanisms, inflammatory responses, genetic factors, and environmental regulation.

2.1.1 MSU crystal deposition and inflammatory activation

The direct trigger of gout is the deposition of MSU crystals within joints and periarticular tissues. MSU crystal formation follows a three-stage kinetic model: nucleation, growth, and aggregation. When serum uric acid (SUA) concentration exceeds 6.8 mg/dL (its saturation solubility), urate precipitates from supersaturated body fluids to form crystals (Bhadouria et al., 2025; Narang and Dalbeth, 2020). Following deposition, these crystals trigger an inflammatory cascade by activating the innate immune system (e.g., macrophages and neutrophils). MSU crystals activate innate immunity via two primary modes. Pattern recognition receptor (PRR)-dependent pathway: cholesterol-rich microdomains exposed on the crystal surface directly bind to Toll-like receptors 2/4 (TLR2/4), thereby initiating the MyD88 signaling cascade (Bhadouria et al., 2025; Narang and Dalbeth, 2020). This leads to NF-κB-mediated expression of pro-inflammatory cytokines (IL-6 and TNF-α), constituting a core mechanism of acute gout attacks. Lysosomal damage pathway: Upon phagocytosis by macrophages, crystals cause lysosomal membrane rupture, releasing cathepsin B, which activates the NLRP3 inflammasome (Kluck et al., 2021; Zhang, 2023). This complex cleaves pro-IL-1β via caspase-1 to generate active IL-1β (a key inflammatory mediator), concurrently inducing gasdermin D-mediated pyroptosis. The integration mechanism of NLRP3 inflammasome activation and the IL-1 signaling pathway, synergistically driving the inflammatory cascade response, is shown in Figure 1. This integrated model reveals the regulatory network from NLRP3 inflammasome activation and effector release to IL-1-mediated amplification of inflammatory cascades.

Figure 1
Diagram illustrating the inflammation process in gout. Uric acid deposition in a foot joint leads to MSU crystals, triggering inflammation. TLR activates MYD88, leading to a cascade involving IRAK, IKK complexes, and NF-κB, resulting in IL1β production. Pyroptosis, inflammasome activation, and mitochondrial ROS further contribute to inflammation, enhancing immune response.

Figure 1. Integrated mechanism of NLRP3 inflammasome activation and IL-1 signaling synergy in driving inflammatory cascades. Note: NLRP3 inflammasome activation occurs via a dual-signal mechanism. Signal 1 (Priming): NF-κB-dependent pathways (e.g., Toll-like receptor activation) induce expressions of NLRP3 and pro-IL-1β. Signal 2 (Activation): MSU crystals trigger NLRP3 oligomerization through K+ efflux, mitochondrial dysfunction, and ROS release. MSU interaction recruits Nek7 kinase, binding NLRP3 to drive inflammasome assembly. Cleavage of gasdermin D, generating pore-forming fragments that induce pyroptosis and inflammatory mediator release. Secreted IL-1β recruits MyD88, IRAK1/2/4, and TRAF6. This activates the IKK complex, leading to NF-κB nuclear translocation and transcription of chemokines/cytokines, establishing a positive feedback loop that amplifies inflammation (created in BioRender. (2025) https://BioRender.com/kp5tvi0).

2.1.2 Genetic predisposition

GWASs have identified multiple gout-associated genetic loci, revealing a dual-pathway “urate metabolism-inflammatory regulation” genetic architecture. This includes urate transporter genes (e.g., ABCG2, URAT1/SLC22A12, and GLUT9/SLC2A9), primarily associated with impaired intestinal/renal uric acid excretion.

Inflammatory regulator genes (e.g., IL-1β and IL-6): primarily linked to inflammasome hyperactivation (Chen et al., 2018; Kawaguchi et al., 2021; Nakayama et al., 2023; Eckenstaler and Benndorf, 2021). Metabolic interaction module genes (e.g., GCKR and PDZK1): involved in dysregulated glucose/lipid metabolism impacting urate homeostasis. Notably, loss-of-function mutations in ABCG2 (e.g., Q141K) impair intestinal urate excretion, thereby significantly increasing the risk of gout (Kawaguchi et al., 2021; Eckenstaler and Benndorf, 2021; Dong et al., 2020). Furthermore, epistasis between PKD2 and ABCG2 further influences serum urate levels, highlighting the importance of polygenic synergy in gout pathogenesis (Dong et al., 2020).

2.1.3 Epigenetic reprogramming

Recent studies have revealed distinct DNA methylation patterns in patients with gout compared to those with hyperuricemic individuals. For instance, the promoter regions of inflammation-related genes (e.g., IL-6 and TNF-α) in patients with gouty arthritis exhibit hypomethylation, potentially promoting inflammation directly and independently of hyperuricemia (Tseng et al., 2020). Additionally, dynamic regulation of histone modifications (e.g., H3K27ac) plays a role. MSU crystals activate the p300/CBP acetyltransferase, inducing H3K27ac enrichment at enhancer regions of inflammatory genes (e.g., CXCL8 and PTGS2) within macrophages, thereby prolonging inflammatory responses (Punzi et al., 2025).

2.1.4 Impaired intestinal and renal excretion

Approximately 90% of patients with gout exhibit impaired uric acid excretion, primarily due to reduced renal urate clearance (Abrahim, 2021). Renal excretion: URAT1 (SLC22A12) is the key urate reabsorption transporter in the renal proximal tubule. Its enhanced function or upregulated expression contributes to hyperuricemia (Nakayama et al., 2023; Abrahim, 2021). Proximal tubular epithelial cells reabsorb urate through coordinated transport mediated by URAT1 (SLC22A12) and GLUT9 (SLC2A9). The URAT1 inhibitor benzbromarone reduces its Vmax, while the SLC2A9 eQTL variant (rs7442295) decreases the fractional excretion of uric acid. Intestinal excretion: ABCG2-mediated intestinal urate excretion accounts for ∼30% of total elimination. ABCG2 dysfunction can induce “secondary hyperuricemia.” Gut microbiota metabolites (e.g., butyrate) upregulate ABCG2 expression by activating PPARγ, suggesting therapeutic potential via microbe–host interaction (Eckenstaler and Benndorf, 2021).

2.1.5 Synergism with metabolic syndrome

Gout is closely associated with metabolic syndrome components, including obesity, insulin resistance, and hypertension (Abrahim, 2021; Pascart and Liote, 2019). An imbalance in adipose tissue-derived adipokines (leptin and adiponectin) may exacerbate gout by inhibiting renal urate excretion and promoting inflammation. An elevated leptin/adiponectin ratio suppresses AMPK phosphorylation, downregulates renal tubular ABCG2 expression, and activates the NLRP3 inflammasome, creating a “metabolic inflammation” microenvironment (Pascart and Liote, 2019; Gong et al., 2020). Furthermore, high fructose intake activates hepatic fructokinase, accelerating ATP degradation into uric acid precursors while inducing insulin resistance. Fructokinase catalyzes fructose conversion to fructose-1-phosphate, consuming ATP to generate AMP, which is subsequently converted to uric acid via the xanthine oxidase (XO) pathway. A single fructose load (>50 g) can increase SUA by 1.5 mg/dL for up to 6 h, establishing a vicious cycle (Pascart and Liote, 2019; Abrahim, 2021).

2.1.6 Transition from asymptomatic hyperuricemia to gout

Only 10%–20% of individuals with hyperuricemia develop gout, implicating critical roles for other regulatory factors (e.g., local tissue microenvironment and crystal-promoting factors) (Ohashi et al., 2024; Zhang, 2023). Studies have shown that asymptomatic individuals with hyperuricemic already exhibit subclinical inflammation (e.g., elevated IL-6 and CRP), whereas acute gout flares are characterized by significant increases in IL-1β and S100A8/A9 levels, which may serve as distinguishing biomarkers (Ohashi et al., 2024; Wu and You, 2023). Thus, the transition from crystal deposition to acute inflammation can be conceptualized as a “two-hit” process: First hit: MSU crystal deposition on cartilage surfaces activates synovial fibroblasts to release CCL2/MCP-1, recruiting monocytes and establishing subclinical inflammation. Second hit: Stimuli such as trauma or hypothermia cause crystal shedding. LL-37 released from neutrophil extracellular traps (NETs) binds crystals, forming pro-inflammatory complexes that trigger an IL-1β storm.

2.1.7 Chronic gout and organ damage

Persistent MSU crystal deposition leads to tophi formation, joint destruction, and chronic kidney disease. Joint damage: MSU crystals within tophi chronically activate macrophages, polarizing them toward an M1 phenotype. These macrophages secrete MMP-13 and ADAMTS-5, degrading type II collagen. Concurrently, TGF-β1-mediated fibrous encapsulation drives joint structural remodeling and cartilage degradation (Towiwat et al., 2019). Renal damage: following endocytosis by renal tubular epithelial cells, MSU crystals activate the TLR9/MyD88 pathway. This promotes NADPH oxidase-derived ROS, which activates the TGF-β/Smad3 signaling pathway, ultimately leading to renal interstitial fibrosis (Wang H. et al., 2018; Luna et al., 2025; Spiga et al., 2017).

2.2 Pathogenesis and pathophysiology of hyperuricemia

Hyperuricemia is the precursor state to gout, but its pathophysiological impact extends far beyond the joints, involving multiple systems, including cardiovascular diseases, chronic kidney disease, and metabolic disorders (Wang Q. et al., 2018; Luna et al., 2025; Spiga et al., 2017). The pathogenesis of hyperuricemia is primarily delineated into two pathophysiological mechanisms: hyperproduction of uric acid and defective renal excretion. Complementary contributing factors encompass genetic predisposition, dysregulated inflammatory pathways, and metabolic comorbidities, which collectively modulate disease expression and progression (Dumusc and So, 2024). Uric acid metabolism and the metabolic–inflammatory axis in hyperuricemia are shown in Figure 2.

Figure 2
Diagram depicting signaling pathways and uric acid metabolism. Left: TLR4 and TNF pathways activate factors like NF-kB and AP1, leading to inflammation and lipid metabolism in the cell nucleus. Right: Uric acid (UA) metabolism, with purine breakdown in the liver, shows excretion via kidneys (ABCG2, OAT1, OAT3) and intestines (ABCG2). Two-thirds of UA from kidneys, one-third from intestines. Arrows indicate pathways and interactions.

Figure 2. Uric acid metabolism and the metabolic–inflammatory axis in hyperuricemia. Note: UA production is catalyzed by XOD, whose activity is modulated by genetic and metabolic factors. UA excretion involves dual regulatory mechanisms: renal homeostasis maintained by URAT1/GLUT9-mediated reabsorption and OAT1-mediated secretion, coupled with intestinal ABCG2-dependent efflux. Metabolic syndrome components dysregulate the ApoA/AdipoQ/FABP4 axis and PPARγ–RXR signaling, promoting hyperuricemia via insulin resistance-induced endogenous purine synthesis and downregulation of OAT1/ABCG2, thereby reducing UA excretion. This initiates a vicious inflammatory cycle: TLR4 recognition of DAMPs activates MyD88-dependent MAP3K8/AKT signaling, inducing NF-κB nuclear translocation and upregulating IL-6/TNF-α; TNF-α further amplifies inflammation via PI3K–AP1 signaling. These cytokines directly activate the renal NLRP3 inflammasome while indirectly reducing UA excretion (through URAT1 downregulation) and promoting crystal deposition via mitochondrial oxidative stress. Impaired intestinal ABCG2 function exacerbates UA retention, reinforcing a pathogenic metabolic–inflammatory axis (created in BioRender. https://BioRender.com/mpuv1n5).

2.2.1 Uric acid overproduction

Abnormal purine metabolism is the primary cause of excessive uric acid production. Uric acid is the final metabolite of purines in humans. Normal adults produce approximately 700 mg of uric acid daily (Jalal et al., 2013). Endogenous purines originate from nucleic acid breakdown of damaged/apoptotic cells, along with adenine and guanine degradation, whereas exogenous purines are primarily derived from dietary sources, including fructose-rich beverages and purine-rich foods (e.g., meat and seafood) (Chaudhary et al., 2013; Weaver, 2019). These exogenous substrates undergo UA biosynthesis in the liver, intestinal, and vascular endothelial compartments. The terminal step of human purine metabolism involves XO-catalyzed oxidation of hypoxanthine to xanthine, followed by further oxidation to uric acid. Circulating UA is distributed via the bloodstream to excretory organs (kidneys and intestines) for elimination (Hille et al., 2014; Wen et al., 2024). As the rate-limiting enzyme in UA synthesis, XO activity is modulated by genetic factors (e.g., HPRT1 mutations) and dietary purine intake. Pharmacological inhibition of XO (e.g., allopurinol) demonstrates marked hypouricemic effects, underscoring the clinical significance of this pathway (Yang et al., 2024). Additionally, accelerated ATP degradation under pathological conditions (e.g., intense exercise and ischemia–reperfusion injury) promotes hypoxanthine accumulation, creating a secondary drive for UA overproduction (Uskudar Cansu et al., 2019).

2.2.2 Impaired renal uric acid excretion

Renal handling of uric acid plays a pivotal role in maintaining systemic urate homeostasis. Approximately 90% of filtered uric acid undergoes reabsorption, while ∼10% is excreted in urine, with glomerular filtration occurring via passive diffusion across capillary endothelial pores (Wen et al., 2024). Renal urate transport involves three sequential processes: glomerular filtration, proximal tubule reabsorption, and active secretion. Dysregulation of urate transporters in the proximal convoluted tubule constitutes the principal determinant of net urinary excretion (Nakayama et al., 2023; Eckenstaler and Benndorf, 2021). In particular, URAT1 (SLC22A12) and OAT10 (SLC22A13) serve as the primary mediators of apical reabsorption, whereas basolateral secretion involves ATP-binding cassette subfamily G member 2 (ABCG2) and sodium-dependent phosphate transporter 1 (NPT1/SLC17A1). Furthermore, metabolic comorbidities such as insulin resistance indirectly enhance urate reabsorption through NHE3-mediated (sodium–hydrogen exchanger 3) tubular alkalinization, creating a mechanistic link between metabolic syndrome and hyperuricemia (Gong et al., 2020; Abrahim, 2021).

2.2.3 Regulatory role of intestinal excretion

Quantitative contribution: Using C14 tracer studies, Sorensen (1965) demonstrated that a significant portion of total body uric acid turnover (∼1/3) is excreted via the intestine (extrarenal elimination pathway) compared to ∼2/3 excreted renally (Du et al., 2024). ABCG2 function: ABCG2, which is highly expressed on the apical membrane of intestinal epithelial cells, plays a critical role in intestinal urate secretion. Its dysfunction can reduce intestinal urate excretion by >50%, significantly elevating serum urate levels. Gut microbiota: Animal models suggest that gut microbiota (e.g., Bifidobacterium) can modulate host urate metabolism by degrading urate precursors, highlighting the potential role of the gut–kidney axis in hyperuricemia (Cabau et al., 2023).

2.2.4 Mechanisms of renal and intestinal urate reabsorption and excretion

Renal and intestinal handling of urate relies on specific transporters to maintain systemic urate levels. These are categorized as follows. Reabsorptive transporters: e.g., glucose transporter 9 (GLUT9/SLC2A9) and urate transporter 1 (URAT1/SLC22A12). Secretory transporters: e.g., organic anion transporters OAT1 and OAT3 (SLC22A6 and SLC22A8), ATP-binding cassette transporter G2 (ABCG2), sodium-dependent phosphate transporter 1 and 4 (NPT1/SLC17A1 and NPT4/SLC17A3), and multidrug resistance protein 4 (MRP4/ABCC4). Renal localization (proximal tubule): reabsorption: URAT1 (apical membrane), GLUT9 (apical and basolateral membranes), and OAT4/10 (mainly apical). Secretion: ABCG2, OAT1/3 (basolateral), MRP4, and NPT1/4 (apical) ensure urate secretion from blood into the tubular lumen (Enomoto et al., 2002; Hu Q. et al., 2024; Yu et al., 2023; Bahn et al., 2008). Intestinal localization: GLUT9 is expressed on the apical and basolateral membranes of murine intestinal epithelial cells. ABCG2 (apical membrane of enterocytes) and NPT1 (apical membrane of small intestinal cells) are also significant intestinal urate transporters (Chen et al., 2019).

2.2.5 Inflammation, oxidative stress, and others

Asymptomatic hyperuricemia exhibits pro-inflammatory and pro-oxidative properties that contribute to multisystem pathology. Elevated SUA activates endothelial NF-κB signaling, upregulating monocyte chemoattractant protein-1 (MCP-1) and intercellular adhesion molecule-1 (ICAM-1) expressions to accelerate atherosclerosis progression (Wang H. et al., 2018; Spiga et al., 2017). Furthermore, microcrystalline urate deposition activates the NLRP3 inflammasome in renal and vascular tissues, exacerbating kidney disease and cardiovascular complications through IL-1β-mediated inflammation (Cabau et al., 2023; Wang Q. et al., 2018).

This metabolic disturbance demonstrates bidirectional interactions with components of metabolic syndrome. Adipose tissue-derived free fatty acids (FFAs) impair renal urate excretion via URAT1 upregulation, while hyperuricemia reciprocally induces insulin resistance and endothelial dysfunction through AMPK inhibition and oxidative stress (Xu et al., 2015). This vicious cycle creates a “metabolic hyperuricemia” phenotype, characterized by stimulated hepatic lipogenesis and dysregulated adipokine levels. Pharmacological contributors include thiazide diuretics (which inhibit renal urate secretion), cyclosporine (which reduces glomerular filtration), and pyrazinamide (which increases purine synthesis), all of which can precipitate secondary hyperuricemia (Zhang K. et al., 2019). Such drug-induced elevations compound the effects of systemic comorbidities, including hypertension, insulin resistance, and atherosclerosis, collectively impairing urate homeostasis through both enhanced production and reduced excretion.

Thus, the pathogenesis of gout is fundamentally driven by hyperuricemia, which serves as both a precursor and a central pathological hub. This condition involves intricate regulation across genetic, epigenetic, metabolic, and immune dimensions, creating a multifactorial disease network. As an independent pathological entity, hyperuricemia extends beyond gout pathogenesis to contribute to systemic comorbidities, including atherosclerosis, chronic kidney disease, and metabolic syndrome. This broad pathophysiological impact underscores the necessity of holistic therapeutic strategies targeting both urate-lowering and anti-inflammatory mechanisms.

3 Diagnosis and treatment of gout and hyperuricemia

3.1 Diagnosis of gout

The diagnosis of gout requires integration of clinical manifestations, laboratory findings, and imaging evidence. According to the classification criteria jointly developed by the American College of Rheumatology (ACR) and the European Alliance of Associations for Rheumatology (EULAR), the identification of MSU crystals in synovial fluid is the diagnostic “gold standard” (Liang et al., 2024; Morlock et al., 2025). For patients where synovial fluid analysis is unfeasible, imaging techniques such as ultrasound and dual-energy computed tomography (DECT) can detect urate deposition, with sensitivities ranging from 15% to 40%. These techniques hold particular value for the early identification of asymptomatic hyperuricemia (Morlock et al., 2025; Dalbeth et al., 2021). The recently developed MSU Crystal Deposition Quantification (MSKF) scoring system, which quantifies urate burden based on ultrasound features (e.g., double-contour sign and tophi), has been validated for distinguishing asymptomatic hyperuricemia from gout. Laboratory diagnosis typically involves SUA levels ≥6.8 mg/dL (404 μmol/L), which represents a critical biochemical prerequisite for gout development (Zhu et al., 2025). However, approximately 50% of patients with chronic hyperuricemia do not progress to clinical gout within 15 years (Singh et al., 2025). Therefore, guidelines emphasize the need to exclude other crystal-induced arthropathies (e.g., pseudogout) and consider the diagnosis in conjunction with typical symptoms such as joint swelling, redness, heat, and pain (Panlu et al., 2024; Morlock et al., 2025).

3.2 Pharmacological treatment of gout

3.2.1 Acute gout flare management

Pharmacotherapy for gout addresses both acute inflammation/pain during flares and long-term urate lowering in the chronic phase. First-line agents for acute flares include the following: NSAIDs: reduce inflammation by inhibiting COX. Potential gastrointestinal and cardiovascular risks must be considered (Bartsch et al., 2025; Yao et al., 2024). Colchicine: exerts anti-inflammatory effects by inhibiting microtubule polymerization and neutrophil chemotaxis (Quintana et al., 2023). A low-dose regimen (e.g., 1.2 mg initially, followed by 0.6 mg after 1 h) is recommended to minimize side effects such as diarrhea. Long-term low-dose colchicine can also be used for flare prophylaxis. Glucocorticoids: effective for acute inflammation, often used orally or via intra-articular injection, especially when NSAIDs or colchicine are contraindicated (Bartsch et al., 2025; Forster, 2024).

3.2.2 ULT for chronic management

Chronic management centers on ULT, using agents from several classes. XO inhibitors (XOIs). Allopurinol: the most commonly used first-line agent, which inhibits uric acid production. Careful monitoring is required due to the risk of hypersensitivity reactions (e.g., Stevens–Johnson syndrome) (Dillman et al., 2024). Febuxostat: a selective XOI suitable for patients intolerant to allopurinol, although associated with an increased risk of cardiovascular events (Quintana et al., 2023). Uricosuric agents. Benzbromarone: promotes renal uric acid excretion by inhibiting tubular reabsorption (primarily via URAT1) (Pino-Zambrano et al., 2024). It is contraindicated in patients with renal stones or significant renal impairment. Uricase agents. Pegloticase: indicated for refractory gout, catalyzes the oxidation of uric acid to highly soluble allantoin, rapidly lowering SUA. Monitoring is required for infusion reactions and the potential development of anti-drug antibodies.

3.2.3 Natural products and plant-derived therapeutics

Research on natural compounds shows promise, although clinical application often faces challenges. Theaflavins: exhibit anti-gout potential by inhibiting inflammatory pathways (e.g., NLRP3 inflammasome) and modulating uric acid metabolism. Limited bioavailability remains a hurdle (Liu G. et al., 2025; Yan et al., 2025). Dioscorea septemloba: network pharmacology analysis suggests that its active components may regulate inflammation and urate metabolism through multiple targets (Liu W. et al., 2025). Taxifolin (dihydroquercetin): shows promise in reducing inflammatory responses and acute flare frequency, particularly when combined with dietary interventions (Piao et al., 2022). In addition, various other natural products have demonstrated efficacy in treating gout and will not be individually listed here.

3.2.4 Exploratory non-pharmacological approaches

Emerging non-pharmacological strategies are under investigation. Probiotics: modulation of the gut microbiota (e.g., using Limosilactobacillus reuteri) has been associated with reduced SUA levels and decreased gout flare frequency (Rodriguez et al., 2023). Transcutaneous auricular vagus nerve stimulation: this technique, which aims to suppress systemic inflammation, may serve as a potential non-pharmacological approach for managing chronic inflammation in the future (Shin et al., 2024). In addition, non-pharmacological interventions for gout, including dietary modifications, weight management, and physical therapies, demonstrate multi-target efficacy by reducing serum uric acid levels, modulating inflammatory pathways, and improving metabolic parameters, thereby decreasing flare frequency, preventing complications, and complementing pharmacotherapy in mild-to-moderate cases.

3.3 Diagnosis of hyperuricemia

Hyperuricemia is defined as SUA levels exceeding 7.0 mg/dL (416 μmol/L) in male individuals and 5.6 mg/dL (333 μmol/L) in female individuals (Wu F. et al., 2024; Tuono et al., 2024). Diagnostic evaluation requires integration of SUA measurements with metabolic syndrome assessment (e.g., obesity and insulin resistance) (Ji et al., 2025). Notably, ultrasonography or DECT reveals urate crystal deposition in 15%–40% of asymptomatic hyperuricemic patients, indicating subclinical gout risk (Xiang et al., 2024; He et al., 2024). However, the demarcation between asymptomatic hyperuricemia and gout remains clinically ambiguous as only approximately 50% of patients with prolonged hyperuricemia develop gout within 15 years (Xiang et al., 2024; Chang et al., 2025). This underscores the necessity of imaging modalities (e.g., DECT) for detecting urate deposition to differentiate subclinical gout (Chang et al., 2025). The definitive diagnostic criterion for gout relies on the identification of MSU crystals in synovial fluid or tissues, with elevated SUA serving solely as a supplementary indicator.

3.4 Pharmacological treatment of hyperuricemia

3.4.1 Conventional agents and mechanisms of action

Pharmacotherapy for hyperuricemia involves multiple mechanisms and drug classes, primarily targeting urate production inhibition, uricosuric effects, and metabolic pathway modulation (Du et al., 2024). XOIs represent the cornerstone of conventional treatment. Allopurinol, a classic XOI, reduces urate synthesis by inhibiting hepatic xanthine oxidoreductase (XOR) (Wu D. et al., 2024). Clinical studies demonstrate that allopurinol combined with febuxostat improves the glomerular filtration rate in non-dialysis CKD patients (Nguyen et al., 2024). Febuxostat, a novel selective XOR inhibitor, exhibits superior efficacy to non-pharmacological interventions in asymptomatic hyperuricemia trials, particularly in ameliorating arterial stiffness assessed using cardio-ankle vascular index (CAVI) (Kawachi et al., 2025). Uricosuric agents, exemplified by benzbromarone, enhance urinary urate excretion via regulation of renal transporters (e.g., OAT1), although hepatotoxicity limits chronic use (Sun et al., 2025).

3.4.2 Natural products and botanical-derived therapies

Natural products are emerging as alternatives to conventional medications. Traditional Chinese medicine formulations, such as Simiao Pills, demonstrate multi-component synergy (phenolic acids, terpenoids, and alkaloids) in mitigating hyperuricemia and renal injury, mediated through anti-inflammatory effects and renal transporter modulation (Li et al., 2022; Zeng et al., 2024). Atractylodes macrocephala exhibits anti-hyperuricemic and anti-inflammatory properties in animal models, reducing IL-1β and TNF-α levels while activating AMPK/SIRT1 signaling and suppressing NF-κB-mediated macrophage polarization toward pro-inflammatory phenotypes (Qian et al., 2023). Polyphenolic compounds, including hazel leaf polyphenols, exert dual inhibitory effects on XOR activity and OAT1/hypoxanthine-guanine phosphoribosyltransferase (HPRT) gene expression, with molecular docking studies predicting PI3K–AKT pathway involvement (Mehmood et al., 2022; Wang R. et al., 2024). Naringenin demonstrates dose-dependent urate-lowering and anti-inflammatory renal effects by inhibiting XOR and modulating inflammatory pathways (Yang Y. et al., 2023). Additionally, isobavachin (derived from Pittosporum species) acts as a human urate transporter 1 (hURAT1) inhibitor, blocking urate reabsorption to reduce SUA levels, akin to the mechanism of tranilast (Luo et al., 2023).

3.4.3 Exploratory non-pharmacological approaches

Non-pharmacological approaches serve as essential adjuncts for hyperuricemia management, particularly in asymptomatic individuals (Paul et al., 2017; Valsaraj et al., 2020). Key dietary modifications include the following. Restriction: limiting alcohol (especially beer/spirits), sugar-sweetened beverages, heavy meat/seafood consumption, and excessive purine intake (reducing SUA by ∼1 mg/dL) (Mitnala et al., 2016). Incorporation: emphasizing cherries, coffee, low-fat dairy, and moderate consumption of protein-rich vegetables (nuts, legumes, and spinach) due to their low urate bioavailability and beneficial fiber (Levy and Cheetham, 2015; Zhang et al., 2012; Park et al., 2016). Supplementation: Vitamin C demonstrates SUA-lowering potential (Peng et al., 2024). Weight management remains fundamental (Du et al., 2024). Although some guidelines support treating asymptomatic hyperuricemia to mitigate hypertension, CKD, and CAD risks, treatment initiation requires individual risk stratification pending further validation of cardiovascular/renal benefits (Waheed et al., 2021; Chales, 2019).

3.5 Limitations of conventional therapeutic paradigms for gout and hyperuricemia

Current clinical management of gout and hyperuricemia faces multifaceted challenges, with therapeutic bottlenecks stemming from the following critical conflicts.

3.5.1 Safety–efficacy trade-offs

Although conventional urate-lowering agents (e.g. XOI allopurinol and URAT1 inhibitor benzbromarone) effectively reduce SUA levels, their clinical utility is constrained by significant organ toxicity risks (Wu F. et al., 2024). Allopurinol-induced hypersensitivity syndrome, characterized by exfoliative dermatitis and liver failure, arises from purine metabolism inhibition (Alotaibi et al., 2024). Benzbromarone exacerbates renal burden through enhanced uricosuria, potentially precipitating tubulointerstitial fibrosis in CKD patients (Kang et al., 2024). These safety concerns necessitate dose limitations, compromising sustained target inhibition efficacy.

3.5.2 Inadequate precision in metabolic regulation

Emerging evidence highlights the multi-organ (hepatic–renal–intestinal) coordination of urate metabolism, yet existing pharmacotherapies predominantly target single nodes (e.g., XO or URAT1). This limitation is particularly pronounced in obesity-associated gout, where XOIs fail to modulate aberrant glycolytic pathways (Elsayed and Elsaid, 2022). Altered activity of glycolytic enzyme GAPDH may promote urate production via perturbation of purine intermediates (e.g., 5-phosphoribosyl-1-pyrophosphate) (Jezewski et al., 2022). Obesity-related insulin resistance activates a glycolysis–purine metabolic axis, driving precursor accumulation that remains unaddressed by current therapies (Panlu et al., 2024). Furthermore, elevated XO activity correlates with adipose tissue dysfunction, potentially exacerbating insulin resistance through ROS-NF-κB signaling (Ali et al., 2024). This “single-target” intervention model demonstrates limited efficacy in obesity-related gout, where metabolic memory effects precipitate rapid urate dysregulation post-treatment cessation. Animal studies demonstrate compensatory upregulation of hepatic purine metabolic enzymes (e.g., PRPP synthetase) following XOI discontinuation, while clinical observations reveal more pronounced SUA rebound in obese gout patients than in non-obese counterparts—a phenomenon potentially mediated by free fatty acid-induced hepatic XO activation from adipose tissue (Cheng L. et al., 2025).

3.5.3 Therapeutic adherence challenges

Chronic conditions (e.g., gout, diabetes, and psychiatric disorders) require prolonged pharmacotherapy (e.g., febuxostat or antipsychotics), yet patient adherence wanes with complex regimens (e.g., twice-daily dosing) (Bartsch et al., 2025; Cai et al., 2024). Long-acting injectable (LAI) formulations reduce the dosing frequency but exhibit variable adherence in real-world practice, with some patients discontinuing therapy due to perceived complexity (Lnu et al., 2024). Concomitant monitoring requirements (SUA levels and liver/renal function) impose additional burdens: frequent phlebotomy provokes patient resistance, while toxicity surveillance (ALT, AST, and creatinine) demands individualized schedules that increase healthcare encounters (Kaur et al., 2024; Wu D. et al., 2024; Park et al., 2024; Boruah et al., 2024). Special populations (e.g., post-renal transplant and CKD patients) require intensified monitoring, further exacerbating compliance challenges (Boruah et al., 2024).

3.6 Innovative potential and clinical translation of DDS

Current advancements in machine learning, bioinformatics, and multi-omics technologies have enabled significant progress in targeted precision interventions for cancer (Shao et al., 2025; Yan et al., 2024). However, precision research on immunometabolic diseases such as gout and hyperuricemia still predominantly relies on DDS. Addressing the limitations of conventional therapies, DDS offers a three-dimensional therapeutic paradigm through interdisciplinary innovations in material science and pharmacokinetics, particularly for gout and hyperuricemia management (Peng et al., 2023).

3.6.1 Organ-specific targeted delivery

Surface-modified nanocarriers (e.g., nanoparticles and liposomes) enable precise drug enrichment in renal proximal tubules or hepatocytes (Peng et al., 2023). Receptor-mediated targeting: mannose receptor-targeted nanocarriers (e.g., liposomes and polymeric nanoparticles) decorated with URAT1-specific ligands demonstrate enhanced tubular accumulation, minimizing gastrointestinal adverse effects associated with systemic drug exposure (Faustino et al., 2023). Trans-physiological barrier delivery: nanoscale carriers overcome limitations of conventional agents in penetrating barriers (e.g., blood–brain barrier and synovial membrane) through size-dependent diffusion and active targeting moieties (e.g., hyaluronic acid-modified silk fibroin nanoparticles), prolonging drug retention at inflammatory sites (Cheng Z. et al., 2025; Garhwal et al., 2024). Toxicity reduction: targeted delivery mitigates hepatotoxicity and hypersensitivity risks inherent to non-selective agents (e.g., allopurinol) while improving therapeutic indices (Ezike et al., 2023).

3.6.2 Spatiotemporally controlled sustained-release platforms

Microsphere/liposome systems: PLGA- or lipid-encapsulated uricase formulations achieve weeks-to-months sustained release, reducing injection frequency and improving adherence in chronic regimens (Ezike et al., 2023). Transdermal systems: novel platforms (e.g., silk fibroin/hyaluronic acid microneedles and chitosan patches) bypass gastrointestinal metabolism, maintaining cutaneous drug concentrations for >7 days—particularly advantageous for obese patients or those with venous access limitations (Harwansh et al., 2023). Environment-responsive release: pH-sensitive nanocarriers (e.g., FeO/chitosan/TiO2 composites) trigger drug release in acidic microenvironments (e.g., inflammation sites and crystal deposits), enhancing local efficacy (Mohamadi Bian and Moghadam, 2025; Liu et al., 2024).

3.6.3 Multimodal synergistic therapeutics

DDS enables “urate-lowering + anti-inflammatory” combinatorial strategies through co-delivery systems. Dual-action nanocarriers: liposomal or polymeric nanoparticles co-encapsulating IL-1β inhibitors and XOIs (e.g., febuxostat) achieve sequential neutralization of inflammatory cytokines and sustained XO inhibition, preventing crystal formation (Liu Y. et al., 2023; Wang et al., 2025). Immunomodulatory integration: nano-formulated PD-1 blockade therapies modulate macrophage polarization and suppress NLRP3 inflammasome activation, mitigating chronic inflammatory damage in gouty arthritis (Liu Y. et al., 2023).

Gout and hyperuricemia management requires integrated approaches addressing metabolic dysregulation and organ-specific damage. Conventional therapies fall short in high-risk populations (e.g., obesity and renal impairment) due to toxicity and adherence limitations. DDS innovations—through pharmacokinetic optimization, precision targeting, and combinatorial strategies—represent transformative solutions to overcome these barriers, offering safer, more effective therapeutic options for complex clinical scenarios.

4 Application of DDS in gout and hyperuricemia

4.1 Introduction to DDSs and its classification in gout/hyperuricemia management

DDSs represent engineered platforms designed to optimize drug pharmacokinetics and biodistribution through advanced carrier technologies (e.g., nanoparticles, colloidal systems, and polymers). Their primary objectives include enhancing bioavailability, improving targeting efficiency, and enabling controlled release profiles to maximize therapeutic efficacy while minimizing off-target toxicity (Garhwal et al., 2024). For instance, nanomedicine-based DDSs (NDDSs) leverage nanoscale architectures to achieve superior target specificity compared to conventional formulations, which often suffer from poor bioavailability and non-selective distribution (Losada-Barreiro et al., 2024). Key DDS design parameters encompass physicochemical properties (e.g., particle size, surface charge, and stability) that directly influence delivery efficiency and safety profiles (Ju and Cho, 2023).

DDS applications span diverse disease domains and administration routes. In oncology, extracellular vesicle-based systems enhance drug penetration into tumor microenvironments, mitigating chemotherapy resistance (Zheng et al., 2024). Ophthalmic lipid-based formulations (e.g., liposomes) improve intraocular drug retention through sustained-release mechanisms (Batur et al., 2024). Pulmonary nanocarriers optimize aerosolized drug deposition in respiratory diseases, while chitosan-based systems enable chronic disease management via mucoadhesive drug delivery (Jeong et al., 2025; Manohar et al., 2025). β-Glucan-functionalized carriers demonstrate efficacy in inflammatory and immune-mediated disorders through biocompatibility-driven targeting (Wu et al., 2023). Collectively, DDS embodies an engineering-driven paradigm focused on precision medicine through tailored carrier design (e.g., polymeric micelles and biomimetic nanodevices).

Recent advances in DDS research for gout and hyperuricemia reflect a paradigm shift toward precision therapy (Karantas et al., 2024). Current classification frameworks categorize DDS into three principal classes: lipid-based, polymer-based, and other systems. These platforms enhance therapeutic performance through targeted delivery, controlled release kinetics, and barrier penetration capabilities. Lipid-based DDSs (LBDDSs) utilize phospholipid bilayer architectures [e.g., liposomes, ethosomes, and solid lipid nanoparticles (SLNs)] to achieve localized drug enrichment in joint cavities or inflammatory foci, with reduced immunogenicity due to biomimetic membrane properties (Seo et al., 2023). Polymer-based DDSs use synthetic (e.g., PLGA) or natural polymers (e.g., chitosan) to fabricate nanocapsules, hydrogels, or microneedle arrays, enabling prolonged drug release profiles particularly advantageous for chronic hyperuricemia management (Behnke et al., 2024). Hybrid systems (e.g., inorganic nanoparticles and EV–mimetic carriers) integrate multifunctional materials or biological motifs to overcome limitations of conventional carriers (Ng et al., 2022).

4.2 LBDDSs for gout and hyperuricemia

LBDDSs represent a class of platforms utilizing lipid materials to enhance drug bioavailability, stability, and targeting efficiency (Savla et al., 2017). Key categories include liposomal systems, SLNs, ethosomal carriers, and self-emulsifying drug delivery systems. These systems encapsulate both hydrophilic and lipophilic drug molecules, improving solubility, absorption, and delivery efficacy across biological barriers. LBDDSs have demonstrated broad applicability in oral, transdermal, topical, and systemic administration for diseases including cancer, autoimmune disorders, and dermatological conditions (Liu X. et al., 2025).

4.2.1 Liposomal systems and functional derivatives

Liposomal systems encompass conventional vesicles and functionalized derivatives (e.g., biomimetic nanoliposomes and elastic liposomes). Composed of phospholipid bilayers, liposomes exhibit biodegradability, nontoxicity, and dual drug-loading capacity for hydrophilic and lipophilic agents (Danaei et al., 2018; Breitsamer and Winter 2019). Their established manufacturing processes enable sustained drug release, active targeting, and combinatorial therapy delivery (Salunkhe et al., 2021). In gout research, liposomal encapsulation of eremantholide C and goyazensolide reduced systemic toxicity while preserving anti-hyperuricemic activity, as evidenced by stabilized SUA reduction and biocompatibility in preclinical models (Ma et al., 2025). For hyperuricemia management, PEGylated liposomal uricase demonstrated enhanced oral bioavailability by protecting enzymes from gastrointestinal degradation and facilitating intestinal epithelial transcytosis (Nishida et al., 1984). Notably, PEGylation prevented immune complex formation and anti-uricase antibody induction in avian models, underscoring its translational potential. Liposomal encapsulation of eremantholide C and goyazensolide in a sesquiterpene formulation mitigates toxicity while maintaining anti-hyperuricemic efficacy through enhanced encapsulation and sustained release, with preclinical evaluation confirming serum uric acid stabilization, excellent biocompatibility, and therapeutic potential for concurrent hyperuricemia/gout management, warranting translational investigation (Cunha Matosinhos et al., 2024).

Biomimetic nanoliposomes represent a cutting-edge subset of LBDDSs. For instance, USM [H]L vesicles integrate encapsulated enzymes to degrade uric acid and hydrogen peroxide, generate photothermal effects for inflammation disruption, repolarize M1 macrophages to M2 phenotypes, and reprogram metabolic/immunological pathways for gout resolution (Chen et al., 2023). Similarly, biomimetic nanovesicles encapsulating uricase achieved 533% and 331% bioavailability improvements compared to native enzymes, with UHLNX formulations demonstrating rapid SUA normalization superior to free uricase (Yang et al., 2020).

Elastic liposomes (ultradeformable liposomes or transfersomes) exhibit exceptional deformability, enabling transdermal penetration through skin barriers (Kumar and Utreja, 2020). Their enhanced permeability profiles reduce systemic side effects in applications such as antihypertensive and topical antibiotic delivery (Yang M. et al., 2019). In gout models, elastic liposomes improved colchicine efficacy by increasing stratum corneum permeation, sustaining drug release to reduce MSU crystal-induced exudate volume and leukocyte infiltration, and inhibiting collagen deposition/inflammatory cell accumulation (Singh et al., 2009).

4.2.2 SLNs and nanostructured lipid carriers

SLNs represent a lipid-based delivery platform characterized by a solid lipid matrix that enables sustained drug release and enhanced chemical stability (Duan et al., 2020). SLNs offer biodegradability, nontoxicity, and superior physical stability compared to liposomes, with simplified manufacturing processes that minimize drug leakage (Duan et al., 2020; Silva et al., 2022). These carriers improve the solubility of poorly water-soluble drugs and accommodate multiple therapeutic agents, making them versatile for oral, transdermal, and topical applications (Mahmoudian et al., 2021). Studies demonstrate that SLNs significantly enhance the solubility and oral bioavailability of [6]-shogaol, a ginger-derived compound. In hyperuricemic/gouty arthritis rat models, SLN-encapsulated [6]-shogaol exhibited superior efficacy compared to the free drug, marked by greater reductions in serum uric acid, IL-1β, and TNF-α levels. Mechanistically, [6]-shogaol reduces urate production through XOD inhibition, while SLN encapsulation provides multi-organ protective effects by mitigating drug degradation and improving biodistribution. These findings position SLNs as promising vehicles for enhancing the anti-gout and urate-lowering properties of hydrophobic compounds (Wang H. et al., 2018).

Nanostructured lipid carriers (NLCs), second-generation lipid systems, incorporate mixed solid–liquid lipid matrices to reduce crystallinity and improve drug loading capacity compared to SLNs (Mahmoudian et al., 2021). This structural optimization minimizes drug expulsion during storage and enhances delivery efficiency (Ashkar et al., 2022). In gout therapy, NLCs demonstrate biocompatibility across administration routes, with controlled-release formulations improving solubility and bioavailability of XOIs (e.g., febuxostat). For instance, NLC-encapsulated febuxostat achieves sustained urate-lowering effects while reducing gastrointestinal side effects associated with oral delivery (Sharma et al., 2022). The hybrid lipid composition of NLCs enables tailored release kinetics, making them advantageous for chronic disease management requiring long-term pharmacotherapy.

4.2.3 Ethosomal systems for transdermal management

Ethosomes represent ethanol-based vesicular carriers characterized by enhanced skin permeation properties, leveraging high ethanol concentrations to disrupt stratum corneum resistance and facilitate drug delivery to epidermal and dermal layers (Kumar and Utreja, 2020). These systems demonstrate superiority in topical anti-inflammatory, antimicrobial, and antihypertensive therapies through increased cutaneous drug deposition (Kumar and Utreja, 2020). Ethosomes’ biodegradability and formulation simplicity enable integration with other carriers for synergistic performance optimization. For instance, a soluble nanoneedle system co-encapsulating colchicine and iguratimod in ethosomes, combined with borneol-modified colchicine ethosomes, achieves dual-mechanism gout treatment. This transdermal platform integrates nanoneedle-mediated osteoclast inhibition/cartilage preservation with borneol-enhanced permeation to suppress MSU crystal-induced inflammation. By stabilizing drug bioavailability while reducing neutrophil activation, pro-inflammatory cytokine secretion, and osteoclast activity, the formulation minimizes gastrointestinal toxicity, offering superior efficacy in managing gout-related pain, joint destruction, and systemic complications compared to conventional oral therapies (Li Z. et al., 2024; Zhang S. et al., 2019).

Ethosomal gels and cataplasms represent semi-solid formulations that enhance topical drug delivery through matrix-controlled release kinetics and prolonged skin retention. As novel transdermal platforms, these systems enable multi-target gout therapy by combining alpha-phellandrene with colchicine (Valsalan Soba et al., 2021). Alpha-phellandrene-loaded cellulose gels block inflammatory mediator synthesis via COX-II and LOX-5 inhibition, while colchicine cataplasms inhibit MSU crystal-induced cytokine release and neutrophil recruitment through transdermal pathways (Valsalan Soba et al., 2021). This dual-component system synergistically reduces inflammation through enzyme inhibition and immunomodulation while significantly mitigating gastrointestinal toxicity and systemic drug exposure, providing a bone-protective strategy with enhanced penetration efficacy for gout-related pain and joint damage.

4.2.4 Self-emulsifying drug delivery systems

Self-emulsifying drug delivery systems (SEDDSs) represent lipid-based formulations that spontaneously form emulsions or microemulsions in gastrointestinal environments, including self-nanoemulsifying DDSs and self-microemulsifying DDSs (SMEDDSs), significantly enhancing the oral bioavailability of lipophilic drugs (Buya et al., 2021). These systems simplify formulation development for poorly soluble compounds by improving intestinal solubility and permeability through emulsion-mediated transport. Lipid composition optimization enables drug stabilization, targeted delivery, and controlled release while minimizing ex vivo degradation (Haddadzadegan et al., 2022; Jampilek and Kralova, 2022). In gout therapy, SEDDSs/SMEDDSs demonstrate significant potential for optimizing pharmacokinetics and enhancing therapeutic indices. Febuxostat-loaded SEDDSs/self-nanoemulsifying lipid transporters (SNELTs) improve XOD inhibitory efficacy by augmenting drug solubility and intestinal absorption, achieving sustained urate-lowering effects with 146.4% relative bioavailability and reduced gastrointestinal toxicity in gouty arthritis models. These formulations optimize pharmacokinetic parameters (elevated Cmax and prolonged AUC) while enabling less frequent dosing, underscoring their clinical utility for managing hyperuricemia-driven inflammation (Rangaraj et al., 2019; Al-Amodi et al., 2020).

In addition, complementary mechanisms provide support for the efficacy of SEDDSs/SMEDDSs in hyperuricemia, as detailed below. Liquiritin–SEDDSs enhance dissolution and multi-organ protection, while morin–phospholipid SMEDDSs synergistically suppress hepatic XOD/XDH expression and restore renal function of uric acid transporters (e.g., mGLUT9 and mURAT1) (Wei et al., 2022). Similarly, 6-shogaol-SMEDDSs achieve 571.18% bioavailability enhancement, targeting XOD inhibition and reducing renal pathology via preferential liver/kidney accumulation (Yang Q. et al., 2019). Isoliquiritigenin– and licochalcone A–SMEDDS further validate this approach, demonstrating XOD suppression, lymphatic transport enhancement, and mitigation of hyperuricemia-induced organ damage (Zhang Y. et al., 2019). Collectively, these systems exemplify how SEDDSs/SMEDDSs enable multifaceted urate control through bioavailability optimization and pathophysiological targeting, offering superior efficacy-to-safety ratios compared to conventional formulations.

In Tables 1 and 2, detailed information on the application of LBDDSs in gout and hyperuricemia is provided. LBDDSs offer transformative potential for gout and hyperuricemia management by enhancing solubility, targeting efficiency, and therapeutic safety. However, critical barriers hinder their clinical translation: (1) regulatory and scalability challenges, such as standardizing sterilization/long-term stability for liposomes/SLNs, require rigorous evaluation of immune responses (e.g., PEGylated liposomal uricase) and cost-effectiveness for advanced systems (biomimetic nanoliposomes and ethosomal gels); (2) preclinical-to-clinical gaps persist in dose optimization (e.g., PK/PD modeling for febuxostat–SEDDSs) and delivery route validation (e.g., human skin studies for elastic liposomes), with limited data on patient compliance and real-world applicability; and (3) technical limitations include liposomal drug leakage, SLN/NLC matrix stability, and ethanol-induced irritation from ethosomes. Future priorities should focus on multi-center trials for safety/PK-PD profiling, biodegradable/low-cost lipid matrix development, and omics-guided biomarker discovery to bridge translational gaps, ensuring that LBDDSs achieve their precision medicine potential in gout and hyperuricemia.

Table 1
www.frontiersin.org

Table 1. Summary of DDS treatment methods for gout.

Table 2
www.frontiersin.org

Table 2. Summary of DDS treatment methods for hyperuricemia.

4.3 Polymeric-based DDSs

Polymeric-based DDSs represent a class of platforms that utilize polymer materials as carriers to enhance drug delivery, playing a pivotal role in the fields of pharmacy, medicine, and healthcare. These systems achieve controlled drug release by modulating the physicochemical properties of polymers, such as solubility, release kinetics, and targeting capabilities, thereby enhancing therapeutic efficacy and reducing adverse effects (Gonzalez et al., 2023). Polymers, which can be natural or synthetic [e.g., chitosan, poly (lactic acid) (PLA), poly (glycolic acid) (PGA)], exhibit excellent biocompatibility and biodegradability, making them suitable for various delivery routes (Harun-Or-Rashid et al., 2023). In the context of gout and hyperuricemia treatment, we categorize these systems into basic polymeric nanocarrier systems, functionalized and composite polymeric carrier systems, hydrogel and fibrous polymeric carrier systems, and microneedle delivery systems.

4.3.1 Basic polymeric nanocarrier systems

Nanoparticle systems use polymeric nanoparticles as core carriers to effectively encapsulate drugs and deliver them to specific sites (e.g., joints and tumor regions) through passive or active targeting mechanisms (Zhao and Zhou, 2022). This design facilitates precise therapeutic delivery, improving bioavailability and reducing systemic toxicity. Common formulations include polymeric nanocapsules, nanospheres, and polymeric nanoparticles, which are utilized in oral or transdermal delivery systems to enhance drug absorption (Singh et al., 2017; Tiwari et al., 2015). Basic polymeric nanocarrier systems significantly enhance the therapeutic efficacy in gout treatment through multi-pathway synergistic mechanisms. For instance, ginsenoside Rb1 nanocapsules orally target inflamed joints, inhibiting the NF-κB/NLRP3 pathway and mitochondrial damage (Liu J. et al., 2020); 15d-PGJ2 nanoformulations block the maturation of various pro-inflammatory cytokines via PPAR-γ-dependent pathways (Ruiz-Miyazawa et al., 2018); and intelligent biomimetic nanosystems combine uricase degradation, near-infrared hyperthermia, and M2 macrophage membrane targeting to achieve dual interventions in uric acid metabolism regulation and inflammatory repair (Xu et al., 2024). These carriers optimize drug delivery efficiency, prolong action duration, and activate multi-target anti-inflammatory pathways, demonstrating superior local efficacy and symptomatic relief compared to conventional administration in rodent and rabbit gout models.

Similarly, basic polymeric nanocarrier systems significantly optimize the therapeutic effects in hyperuricemia through multi-mechanism synergies. Scopoletin and pinocembrin, when encapsulated in polymeric micelles, exhibit urate-lowering effects by inhibiting xanthine oxidase activity and promoting uric acid excretion, respectively, while significantly improving renal injury protection (Zeng et al., 2020; Rong et al., 2022; Zeng et al., 2017). Uricase nanocapsules extend the catalytic activity cycle of the enzyme, converting uric acid into readily excretable allantoin, whereas allopurinol nanoformulations enhance the inhibition of uric acid synthesis and the prevention of kidney stones through targeted delivery (Kandav et al., 2020; Xiong et al., 2016). Furthermore, these nanosystems prolong uricase efficacy through immunomodulation, collectively highlighting the pivotal role of nanocarriers in enhancing drug bioavailability, targeting, and multi-pathway synergistic therapy.

4.3.2 Functionalized and composite polymeric carrier systems

Polymeric carriers are often engineered through surface modification or multifunctional design to enable drug loading, controlled release, and targeted delivery, thereby enhancing therapeutic efficacy while minimizing toxicity and adverse effects (Lima and Reis, 2021). These systems manifest in diverse forms, including nanoparticles, conjugates, and composite systems, offering advantages such as high drug-loading capacity, sustained release profiles, and reduced immunogenicity (Gulyaev et al., 2025). For instance, PDA–PEG and PDA–PEG–TPP nanoparticles (where TPP denotes triphenylphosphine) facilitate targeted delivery to the nucleus and mitochondria, enhancing drug accumulation in cancer cells and overcoming drug resistance (Li et al., 2017). Studies demonstrate that PDA–PEG–TPP–DOX exhibits superior mitochondrial targeting capability both in vitro and in vivo. Furthermore, these systems synergistically degrade uric acid via platinum-catalyzed reactions, scavenge ROS using polydopamine, and enhance mitochondrial repair/anti-inflammatory effects through near-infrared-II photothermal therapy-mediated suppression of NF-κB signaling and pro-inflammatory cytokine release (Zhao et al., 2024).

The BmK9–uricase nanoparticle represents a composite carrier system based on electrostatic complexation between neutral scorpion toxin BmK9 and uricase. This Nplex platform enables synergistic urate-lowering and anti-inflammatory effects via BmK9-mediated cytokine modulation. The nanocomposite architecture ensures sustained drug release, prolonged half-life, and enhanced tissue penetration, collectively mitigating gouty inflammation and preventing urate crystal-induced nephropathy (Han et al., 2021).

Additionally, PEG–drug conjugates are prevalent functionalized carriers where PEG modifies therapeutic agents (e.g., uricase) to improve pharmacokinetics and reduce immunogenicity (Ozer et al., 2022). For example, uricase–PEG conjugates diminish immunogenicity and extend half-life. Similarly, PEGylated IL-1Ra chimeric protein nanoparticles prolong half-life, enabling sustained blockade of the IL-1β pathway and inhibition of MSU crystal-induced inflammation. In clinical settings, pegloticase and SEL-212 formulations, combined with immunomodulators (e.g., methotrexate/sirolimus), suppress anti-drug antibody formation to maintain uricase activity, achieving long-term uric acid control and tophus resolution. Furthermore, sodium-containing formulations should be judiciously avoided in topical preparations, while fully optimized delivery systems are indispensable for enhancing transdermal NSAID penetration, enabling precise targeting to inflammatory foci, and achieving synergistic therapeutic outcomes through dual anti-inflammatory and crystal-dissolving mechanisms (Hooper and He, 2022). By extending drug action cycles, inducing immune tolerance, and activating multi-target synergistic mechanisms, these carrier systems offer highly efficient and safe delivery strategies for gout management.

4.3.3 Hydrogel and fibrous polymeric carrier systems

Hydrogel and fibrous polymeric carrier systems enable precise targeting and multi-mechanistic synergies in gout therapeutics through localized delivery, sustained release, and environmental responsiveness, thereby enhancing efficacy while reducing adverse effects. For instance, composite nanofibers composed of gelatin, chitosan, collagen, or sodium alginate can be fabricated via electrospinning to create porous scaffolds that mimic the extracellular matrix, promoting cell adhesion and proliferation (e.g., spindle-shaped human dermal fibroblasts on gelatin/chitosan scaffolds) (Pezeshki-Modaress et al., 2018). Electrospun poly (lactic-co-glycolic acid)/gelatin composite fibers loaded with luteolin achieve sustained intra-articular release, suppressing MSU-induced TNF-α, IL-1β, and IL-6 production while inhibiting xanthine oxidase activity for long-term recurrence prevention in rabbit models (Wang et al., 2016).

The fundamental attributes of hydrogels lie in their three-dimensional hydrophilic polymer networks, which exhibit biocompatibility, biodegradability, and high drug-loading capacity, enabling on-demand drug release in response to environmental stimuli (e.g., pH and temperature) (Mathew et al., 2018). Transdermal febuxostat-loaded hydroxypropyl methylcellulose hydrogels enhance drug permeability via ethosomal formulations, enabling sustained urate-lowering effects in gouty rats (El-Shenawy et al., 2019). pH-responsive CMCS@SAG hydrogel microspheres selectively release Gas6 in acidic joint microenvironments, promoting M2 macrophage polarization and mitochondrial stabilization to resolve acute gouty inflammation (Chen F. et al., 2025). Additionally, PEG–PAEU–ABP hybrid hydrogels loaded with uricase–human serum albumin effectively stabilize serum uric acid levels in hyperuricemic mice, offering prolonged nephron protection against hyperuricemic damage through extended and sustained release mechanisms (Cho et al., 2020). Collectively, these systems exemplify spatially controlled drug release, microenvironment-responsive activation, and combinatorial anti-inflammatory/urate-regulating strategies for advanced gout therapy.

4.3.4 Microneedle delivery systems

Microneedles represent a minimally invasive drug delivery platform composed of microscale needles (height: 10–2000 μm; width: 10–50 µm) designed to penetrate the stratum corneum barrier and create transient microchannels for painless, transdermal drug administration into the dermis (Hao et al., 2017). Classified by morphology and drug release mechanisms, microneedle variants include solid, coated, dissolving, hollow, and hydrogel-forming types (Kumar and Shukla, 2024). For example, solid microneedles use a “poke-and-patch” strategy, whereas dissolving microneedles directly release encapsulated drugs upon dissolution within the skin (He et al., 2019). Microneedle technology offers advantages such as enhanced bioavailability, reduced pain, improved patient compliance, and ease of operation compared to conventional injections (Zhao et al., 2020). Microneedle delivery systems have emerged as transformative tools for gout therapy, enabling localized and sustained drug release while minimizing systemic toxicity. Colchicine-loaded hydrogel microneedles demonstrate prolonged anti-inflammatory efficacy by downregulating pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) and reducing neutrophil infiltration in gouty joints, as evidenced in rat arthritis models (Jiang et al., 2023). Dissolvable silk fibroin microneedles enhance drug delivery efficiency through robust mechanical penetration, ensuring targeted suppression of cytokine cascades and edema in acute gouty inflammation, with reduced systemic exposure compared to oral administration (Liao et al., 2023).

Microneedle-based platforms address critical limitations of conventional oral therapies for hyperuricemia, such as hepatic first-pass metabolism and poor bioavailability. Transdermal microneedles loaded with allopurinol achieve sustained urate-lowering effects by controlling drug release kinetics while minimizing gastrointestinal side effects observed with oral formulations (Chen et al., 2021). Advanced systems integrating uricase and horseradish peroxidase within CaHPO4 nanoflowers, coupled with hyaluronic acid microneedles, enhance enzyme stability and catalytic activity, effectively reducing serum uric acid levels by 30% in murine models (Hao et al., 2019). Collectively, these platforms underscore the potential of microneedles to improve treatment adherence, efficacy, and safety in hyperuricemia management.

Polymeric drug delivery systems (PDDS) offer transformative potential for gout and hyperuricemia through controlled release, targeted delivery, and multi-mechanistic synergies. The specific details of this part can be found in Tables 1 and 2, which provide a comprehensive overview of the application of DDSs in the treatment of gout and hyperuricemia for this category. However, critical barriers impede clinical translation. (1) Regulatory and scalability gaps: standardized protocols for polymeric carrier scalability, sterilization, and long-term stability are lacking, with advanced systems (e.g., PEGylated uricase and biomimetic nanosystems) requiring rigorous immunogenicity (anti-drug antibodies) and cost-effectiveness evaluations. (2) Preclinical–clinical disconnect: dosing regimens (e.g., colchicine microneedles) lack human PK/PD validation, while transdermal hydrogels need human skin permeability studies to address species-specific stratum corneum variability. (3) Technical limitations: polymeric nanocarriers suffer from premature drug leakage; hydrogels require mechanical strength optimization; and microneedles lack large-scale manufacturing validation and patient compliance data. Future efforts must prioritize multi-center trials for safety/PK profiling, biodegradable polymers with tunable degradation, and multi-omics biomarkers for personalized therapy. Bridging these gaps will facilitate the adoption of PDDS in precision gout management.

4.4 Other DDSs

Beyond conventional nanocarriers, emerging DDS platforms demonstrate unique advantages in managing gout and hyperuricemia through tailored mechanisms. For gout, low-Zn2+-doped zinc ferrite nanoparticles exhibit dual anti-inflammatory and antioxidant effects by modulating NF-κB/NLRP3 signaling and activating the Nrf2 pathway, thereby reducing joint inflammation and oxidative stress in murine models (Zhang et al., 2025). Electrically responsive polypyrrole bilayer devices enable precise, wireless-controlled transdermal delivery of sodium salicylate, minimizing systemic NSAID toxicity while maintaining localized anti-inflammatory efficacy (Liu Y. et al., 2020). Additionally, Pt/CeO2 nanozymes mimic natural uricase/catalase activity through self-cascade uric acid degradation and H2O2 scavenging, offering a biomimetic approach to mitigate urate crystal-induced oxidative damage in vitro (You and Chen, 2024).

In hyperuricemia management, synthetic gene networks integrating HucR-based biosensors and secreted Aspergillus flavus urate oxidase achieve self-regulated urate degradation, dynamically adjusting enzyme expression to maintain physiological UA levels while avoiding excessive urate depletion (Kemmer et al., 2010). Artificial organelles (Uri/Cat@ArtPC) leverage ε-polylysine/DNA phase separation to co-encapsulate uricase and catalase, enabling synergistic urate conversion and H2O2 neutralization, with enhanced renal targeting and catalytic durability (Hu H. et al., 2024). These platforms collectively advance therapeutic precision by integrating biosensing, self-regulated enzyme activity, and biomimetic catalysis, addressing critical limitations of traditional urate-lowering therapies. However, due to the overlapping and distinct mechanisms underlying DDS applications in gout and hyperuricemia, published research to date provides comprehensive analyses, as summarized in Tables 1 and 2.

Emerging DDSs for gout and hyperuricemia, including zinc ferrite nanoparticles and synthetic gene networks, demonstrate innovative mechanisms such as self-regulated enzyme activity and biomimetic catalysis. Tables 1, 2 elaborate on the detailed application of the DDS in gout and hyperuricemia, respectively. However, clinical translation is hindered by regulatory/scalability challenges, cost barriers, preclinical–clinical translation gaps, and technical limitations (e.g., wireless control precision and in vivo stability). Future priorities include optimizing biodegradability, conducting pilot trials, and integrating multi-omics data to advance precision therapy.

4.5 Artificial intelligence DDS platform

Although AI-based DDSs remain underexplored in gout and hyperuricemia management, their transformative potential is evident. By integrating bioinformatics data with machine learning algorithms, AI can accelerate target discovery, predict pharmacokinetic profiles, and construct intelligent databases for real-time analysis of multi-omics data, thereby enabling personalized delivery strategies (Hassanzadeh et al., 2019). Key advantages include optimizing nanorobot design, predicting trans-biological barrier behavior, and dynamically adjusting delivery parameters via real-time feedback loops (Rai et al., 2023). Future directions may involve developing urate-sensitive smart carriers for targeted renal or joint delivery, utilizing pH-responsive/enzyme-triggered mechanisms to achieve spatiotemporal control of urate-lowering agents. Additionally, AI-driven disease progression models could optimize delivery timing, while metabolic profiling data may guide the development of systems targeting renal urate transporters (e.g., mURAT1), enhancing intervention precision for chronic metabolic disorders. These innovations hold promise to elevate therapeutic efficacy and patient-specific outcomes in gout management.

5 Conclusion and prospect

In conclusion, this review underscores the revolutionary potential of DDSs in transforming gout and hyperuricemia management through enhanced precision, efficacy, and patient compliance. Modern DDSs—including lipid-based, polymeric, and stimuli-responsive platforms—have demonstrated superiority over conventional therapies by optimizing pharmacokinetics, minimizing off-target effects, and enabling targeted delivery to pathological sites (e.g., renal tubules and inflamed joints). Notable advancements, such as uricase-loaded nanocarriers with reduced immunogenicity and pH-responsive hydrogels for sustained urate control, exemplify the shift toward intelligent, patient-centric designs.

However, critical limitations must be acknowledged. First, clinical validation remains inadequate: although preclinical data highlight the promise of multimodal therapies (e.g., co-delivering urate-lowering agents and anti-inflammatory drugs), robust clinical evidence is lacking. Second, safety and ethical concerns persist, particularly for injectable systems such as PEGylated uricase, where immunogenicity and long-term tissue accumulation necessitate rigorous risk–benefit analyses. Third, technical barriers—such as the scalability of nanofabrication processes, the cost-effectiveness of advanced materials, and heterogeneity in patient response—hinder translational progress.

To address these gaps, future research should prioritize the following. (1) Multifunctional DDS development: integrating real-time monitoring (e.g., biosensors for urate levels) with adaptive release mechanisms to enable personalized dose adjustment. (2) Preclinical-to-clinical translation: conducting phase I/II trials to validate efficacy, safety, and pharmacokinetic profiles of emerging platforms (e.g., artificial organelles and nanozymes). (3) Ethical and regulatory frameworks: establishing guidelines for immunogenicity testing, long-term safety surveillance, and patient consent protocols for experimental therapies. (4) Interdisciplinary collaboration: leveraging AI and omics technologies to predict therapeutic outcomes, optimize material properties, and identify biomarkers for stratified patient populations.

The future of DDSs for gout and hyperuricemia hinges on three interconnected directions. First, the integration of advanced biomaterials with artificial intelligence could enable self-regulating systems capable of dynamic dose adjustment based on individual metabolic fluctuations. Second, multimodal therapies combining urate-lowering agents, anti-inflammatory cytokines, and immunomodulators within a single platform may synergize to halt disease progression while minimizing systemic side effects. Third, decentralized manufacturing technologies, such as 3D bioprinting and microfluidic synthesis, could democratize access to customized DDSs, particularly in resource-limited settings.

The evolution of DDSs from passive carriers to intelligent, patient-centric platforms signifies promising progress in gout and hyperuricemia management, yet substantial challenges in clinical validation, scalability, and long-term safety persist, underscoring the need for sustained interdisciplinary efforts to fully realize their therapeutic potential. Realizing this potential requires addressing critical gaps: although preclinical studies highlight innovative mechanisms (e.g., multimodal therapies and stimuli-responsive release), robust clinical validation remains scarce, necessitating phase I/II trials to confirm efficacy and safety. Ethical and technical challenges—such as immunogenicity risks in injectable formulations (e.g., PEGylated uricase), long-term biocompatibility of advanced materials, and scalability of nanofabrication processes—demand rigorous surveillance frameworks and cost-effective manufacturing strategies. Ultimately, the field’s progression hinges on interdisciplinary collaboration to integrate adaptive technologies (e.g., AI-driven real-time monitoring), establish standardized safety protocols, and prioritize patient-centered design. By confronting these barriers, DDS can transition from conceptual breakthroughs to clinically validated solutions, offering safer, more equitable, and globally accessible therapeutic strategies to reduce the socioeconomic burden of gout and hyperuricemia.

Author contributions

ZP: Investigation, Writing – original draft. FM: Investigation, Writing – original draft. QD: Investigation, Writing – original draft. YH: Writing – original draft. DM: Writing – original draft. YL: Writing – original draft. WY: Writing – original draft. JP: Funding acquisition, Writing – review and editing. XW: Conceptualization, Supervision, Writing – review and editing. NL: Writing – review and editing, Project administration.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. This work was financially supported by the Yunnan Clinical Research Center for Rheumatism in Traditional Chinese Medicine (202405AJ310004), the Yunnan Science and Technology Talent and Platform Program (202105AG070012), the Yunnan University of Chinese Medicine–Hospital Joint Fund Projects (XYLH2024042 and XYLH2024030), the Science Research Foundation of Education Department of Yunnan Province (2025Y0638 and 2024Y380), the Expert Workstation of Zhangxuan in Yunnan Province (202305AF150175), the Funding of Yunnan Applied Basic Research Projects-Union Foundation (202401AZ070001-052) and the High level Key Discipline of TCM Construction Project, Rheumatology, of National Administration of Traditional Chinese Medicine (zyyzdxk-2023189).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abrahim, M. (2021). Non-alcoholic fatty kidney disease (NAFKD): a novel disease that explains the etiology of hyperuricemia and gout in metabolic syndrome. Med. Hypotheses. 146, 110461. doi:10.1016/j.mehy.2020.110461

PubMed Abstract | CrossRef Full Text | Google Scholar

Al-Amodi, Y. A., Hosny, K. M., Alharbi, W. S., Safo, M. K., and El-Say, K. M. (2020). Investigating the potential of transmucosal delivery of febuxostat from oral lyophilized tablets loaded with a self-nanoemulsifying delivery system. Pharmaceutics 12 (6), 534. doi:10.3390/pharmaceutics12060534

PubMed Abstract | CrossRef Full Text | Google Scholar

Ali, N., Taher, A., Islam, N., Sarna, N. Z., and Islam, F. (2024). Evaluation of the relationship between xanthine oxidase activity and metabolic syndrome in a population group in Bangladesh. Sci. Rep. 14 (1), 20380. doi:10.1038/s41598-024-71733-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Alotaibi, T., Bjazevic, J., Kim, R., Gryn, S., Sultan, N., Dresser, G., et al. (2024). Allopurinol hypersensitivity syndrome raising awareness of an uncommon but potentially serious adverse event among kidney stone patients. CUAJ-Can. Urol. Assoc. J. 18 (5), E167–E172. doi:10.5489/cuaj.8685

PubMed Abstract | CrossRef Full Text | Google Scholar

Ashkar, A., Sosnik, A., and Davidovich-Pinhas, M. (2022). Structured edible lipid-based particle systems for oral drug-delivery. Biotechnol. Adv. 54, 107789. doi:10.1016/j.biotechadv.2021.107789

PubMed Abstract | CrossRef Full Text | Google Scholar

Bahn, A., Hagos, Y., Reuter, S., Balen, D., Brzica, H., Krick, W., et al. (2008). Identification of a new urate and high affinity nicotinate transporter, hOAT10 (SLC22A13). J. Biol. Chem. 283 (24), 16332–16341. doi:10.1074/jbc.M800737200

PubMed Abstract | CrossRef Full Text | Google Scholar

Bartsch, V., Standfest, K., and Hueber, A. (2025). Gout: from the diagnosis to guideline-based treatment. Z. Gerontol. Geriatr. 58 (2), 137–146. doi:10.1007/s00391-025-02416-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Batur, E., Ozdemir, S., Durgun, M. E., and Ozsoy, Y. (2024). Vesicular drug delivery systems: promising approaches in ocular drug delivery. Pharmaceuticals 17 (4), 511. doi:10.3390/ph17040511

PubMed Abstract | CrossRef Full Text | Google Scholar

Behnke, M., Holick, C. T., Vollrath, A., Schubert, S., and Schubert, U. S. (2024). Knowledge-based design of multifunctional polymeric nanoparticles. Handb. Exp. Pharmacol. 284, 3–26. doi:10.1007/164_2023_649

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhadouria, V. S., Verma, S., Agarwal, C., and Sharma, D. S. (2025). From patents to progress: unraveling gout's journey through clinical trials and advancements. Rev. Recent Clin. Trials 20 (2), 96–112. doi:10.2174/0115748871308473240926044126

PubMed Abstract | CrossRef Full Text | Google Scholar

Boruah, P., Ruram, A., Baruah, A. J., Nongtdu, B., Barman, B., Sahoo, D. P., et al. (2024). Association of serum uric acid level and thyroid function in chronic kidney disease: a hospital-based cross-sectional study from northeast India. Cureus 16 (9), e69392. doi:10.7759/cureus.69392

PubMed Abstract | CrossRef Full Text | Google Scholar

Botson, J. K., Obermeyer, K., Lamoreaux, B., Zhao, L., Weinblatt, M. E., and Peterson, J. (2022). Improved joint and patient-reported health assessments with pegloticase plus methotrexate co-therapy in patients with uncontrolled gout: 12-Month exploratory outcomes of the MIRROR open-label trial. Arthritis Res. Ther. 24 (1), 281. doi:10.1186/s13075-022-02979-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Breitsamer, M., and Winter, G. (2019). Vesicular phospholipid gels as drug delivery systems for small molecular weight drugs, peptides and proteins: state of the art review. Int. J. Pharm. 557, 1–8. doi:10.1016/j.ijpharm.2018.12.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Buya, A. B., Witika, B. A., Bapolisi, A. M., Mwila, C., Mukubwa, G. K., Memvanga, P. B., et al. (2021). Application of lipid-based nanocarriers for antitubercular drug delivery: a review. Pharmaceutics 13 (12), 2041. doi:10.3390/pharmaceutics13122041

PubMed Abstract | CrossRef Full Text | Google Scholar

Cabau, G., Gaal, O., Badii, M., Nica, V., Mirea, A., Hotea, I., et al. (2023). Hyperuricemia remodels the serum proteome toward a higher inflammatory state. iScience 26 (10), 107909. doi:10.1016/j.isci.2023.107909

PubMed Abstract | CrossRef Full Text | Google Scholar

Cai, C., Kozma, C., Patel, C., Benson, C., Yunusa, I., Zhao, P., et al. (2024). Adherence, health care utilization, and costs between long-acting injectable and oral antipsychotic medications in South Carolina medicaid beneficiaries with schizophrenia. J. Manag. Care Spec. Pharm. 30 (6), 549–559. doi:10.18553/jmcp.2024.30.6.549

PubMed Abstract | CrossRef Full Text | Google Scholar

Chales, G. (2019). How should we manage asymptomatic hyperuricemia? Jt. Bone Spine 86 (4), 437–443. doi:10.1016/j.jbspin.2018.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Chang, H., Lv, J., Zheng, Y., Li, D., and Li, Y. (2025). The diagnostic value of serum MCP-1 combined with OPN detection for early renal injury in gout patients. Int. J. Gen. Med. 18, 1423–1429. doi:10.2147/IJGM.S508220

PubMed Abstract | CrossRef Full Text | Google Scholar

Chaudhary, K., Malhotra, K., Sowers, J., and Aroor, A. (2013). Uric acid - key ingredient in the recipe for cardiorenal metabolic syndrome. Cardiorenal Med. 3 (3), 208–220. doi:10.1159/000355405

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, C., Tseng, C., Yen, J., Chang, J., Chou, W., Chu, H., et al. (2018). ABCG2 contributes to the development of gout and hyperuricemia in a genome-wide association study. Sci. Rep. 8 (1), 3137. doi:10.1038/s41598-018-21425-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Chen, D., Chen, L., Liu, J., Vaziri, N. D., Guo, Y., et al. (2019). Microbiome-metabolome reveals the contribution of gut-kidney axis on kidney disease. J. Transl. Med. 17 (1), 5. doi:10.1186/s12967-018-1756-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J., Liu, X., Liu, S., He, Z., Yu, S., Ruan, Z., et al. (2021). Fabrication and characterization of dissolving microneedles for transdermal drug delivery of allopurinol. Drug Dev. Ind. Pharm. 47 (10), 1578–1586. doi:10.1080/03639045.2022.2027959

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, R., Yang, J., Wu, M., Zhao, D., Yuan, Z., Zeng, L., et al. (2023). M2 macrophage hybrid membrane-camouflaged targeted biomimetic nanosomes to reprogram inflammatory microenvironment for enhanced enzyme-thermo-immunotherapy. Adv. Mat. 35 (39), e2304123. doi:10.1002/adma.202304123

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, F., Zhou, Y., Du, X., Lin, S., Wan, W., Liu, Y., et al. (2025). In situ sustained and pH-responsive Gas6-loaded microspheres for alleviating acute gouty arthritis by immunoregulation. Colloid Surf. B-Biointerfaces 254, 114809. doi:10.1016/j.colsurfb.2025.114809

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Z., Guo, Q., Zhang, Y., Chen, L., Li, P., Cheng, W., et al. (2025). A systems immunology perspective on gout pathogenesis and its precision-targeted treatment strategies. Front. Immunol. 16, 1615914. doi:10.3389/fimmu.2025.1615914

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, L., Zhang, Y., Xu, Q., Li, Z., Liu, Z., and Dai, F. (2025). Hyaluronic acid/silk fibroin nanoparticles loaded with methotrexate for topical treatment of psoriasis. Int. J. Pharm. X. 9, 100312. doi:10.1016/j.ijpx.2024.100312

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, Z, Z., Xu, X., Qi, H., Li, X., Li, Y., Jiang, C., et al. (2025). Obesity reduces the urate-lowering efficacy among patients with primary gout: a prospective cohort study. RHEUMATOLOGY 64 (6), 3500–3508. doi:10.1093/rheumatology/keaf076

PubMed Abstract | CrossRef Full Text | Google Scholar

Cho, J., Kim, S. H., Yang, B., Jung, J. M., Kwon, I., and Lee, D. S. (2020). Albumin affibody-outfitted injectable gel enabling extended release of urate oxidase-albumin conjugates for hyperuricemia treatment. J. Control. Release. 324, 532–544. doi:10.1016/j.jconrel.2020.05.037

PubMed Abstract | CrossRef Full Text | Google Scholar

Cumpelik, A., Ankli, B., Zecher, D., and Schifferli, J. A. (2016). Neutrophil microvesicles resolve gout by inhibiting C5a-mediated priming of the inflammasome. Ann. Rheum. Dis. 75 (6), 1236–1245. doi:10.1136/annrheumdis-2015-207338

PubMed Abstract | CrossRef Full Text | Google Scholar

Cunha Matosinhos, R., Frezard, F., Mendes Silva Araujo, S., Magalhaes Barbosa, A., de Souza, I. F., de Souza Filho, J. D., et al. (2024). Development and characterization of liposomal formulations containing sesquiterpene lactones for the treatment of chronic gout. Sci. Rep. 14 (1), 6991. doi:10.1038/s41598-024-57663-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Dalbeth, N., Gosling, A. L., Gaffo, A., and Abhishek, A. (2021). Gout. Lancet 397 (10287), 1843–1855. doi:10.1016/S0140-6736(21)00569-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Danaei, M., Dehghankhold, M., Ataei, S., Hasanzadeh Davarani, F., Javanmard, R., Dokhani, A., et al. (2018). Impact of particle size and polydispersity index on the clinical applications of lipidic nanocarrier systems. Pharmaceutics 10 (2), 57. doi:10.3390/pharmaceutics10020057

PubMed Abstract | CrossRef Full Text | Google Scholar

Deng, X., He, D., Xiong, H., Zhou, Y., and Zhang, J. (2015). The pharmacokinetics and pharmacodynamics of intravenous uricase multivesicular liposomes. Sichuan Da Xue Xue Bao Yi Xue Ban. 46 (5), 688–691. doi:10.13464/j.scuxbyxb.2015.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Dillman, K. M., Hawkins, A. M., Ragland, A. R., Wester, G. C., Greene, D. R., Varrassi, G., et al. (2024). Allopurinol: clinical considerations in the development and treatment of Stevens-Johnson syndrome, toxic epidermal necrolysis, and other associated drug reactions. Cureus 16 (7), e64654. doi:10.7759/cureus.64654

PubMed Abstract | CrossRef Full Text | Google Scholar

Dong, Z., Zhou, J., Jiang, S., Li, Y., Zhao, D., Yang, C., et al. (2020). Epistatic interaction between PKD2 and ABCG2 influences the pathogenesis of hyperuricemia and gout. Hereditas 157 (1), 2. doi:10.1186/s41065-020-0116-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Du, L., Zong, Y., Li, H., Wang, Q., Xie, L., Yang, B., et al. (2024). Hyperuricemia and its related diseases: mechanisms and advances in therapy. Signal Transduct. Target. Ther. 9 (1), 212. doi:10.1038/s41392-024-01916-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Duan, Y., Dhar, A., Patel, C., Khimani, M., Neogi, S., Sharma, P., et al. (2020). A brief review on solid lipid nanoparticles: part and parcel of contemporary drug delivery systems. RSC Adv. 10 (45), 26777–26791. doi:10.1039/d0ra03491f

PubMed Abstract | CrossRef Full Text | Google Scholar

Dumusc, A., and So, A. (2024). Advances in treatment of hyperuricemia and gout. Ther. Umsch. 81 (5), 168–171. doi:10.23785/TU.2024.05.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Eckenstaler, R., and Benndorf, R. A. (2021). The role of ABCG2 in the pathogenesis of primary hyperuricemia and Gout-An update. Int. J. Mol. Sci. 22 (13), 6678. doi:10.3390/ijms22136678

PubMed Abstract | CrossRef Full Text | Google Scholar

El-Shenawy, A. A., Abdelhafez, W. A., Ismail, A., and Kassem, A. A. (2019). Formulation and characterization of nanosized ethosomal formulations of antigout model drug (febuxostat) prepared by cold method: in Vitro/Ex Vivo and in vivo assessment. AAPS PharmSciTech 21 (1), 31. doi:10.1208/s12249-019-1556-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Elsayed, S., and Elsaid, K. A. (2022). Protein phosphatase 2A regulates xanthine oxidase-derived ROS production in macrophages and influx of inflammatory monocytes in a murine gout model. Front. Pharmacol. 13, 1033520. doi:10.3389/fphar.2022.1033520

PubMed Abstract | CrossRef Full Text | Google Scholar

Enomoto, A., Kimura, H., Chairoungdua, A., Shigeta, Y., Jutabha, P., Cha, S. H., et al. (2002). Molecular identification of a renal urate anion exchanger that regulates blood urate levels. Nature 417 (6887), 447–452. doi:10.1038/nature742

PubMed Abstract | CrossRef Full Text | Google Scholar

Ezike, T. C., Okpala, U. S., Onoja, U. L., Nwike, C. P., Ezeako, E. C., Okpara, O. J., et al. (2023). Advances in drug delivery systems, challenges and future directions. Heliyon 9 (6), e17488. doi:10.1016/j.heliyon.2023.e17488

PubMed Abstract | CrossRef Full Text | Google Scholar

Faustino, C., Duarte, N., and Pinheiro, L. (2023). Triterpenes drug delivery systems, a modern approach for arthritis targeted therapy. Pharmaceuticals 17 (1), 54. doi:10.3390/ph17010054

PubMed Abstract | CrossRef Full Text | Google Scholar

Forster, A. (2024). Therapy of gout in 2024. Ther. Umsch. 81 (5), 160–163. doi:10.23785/TU.2024.05.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Garhwal, A., Kendya, P., Soni, S., Kori, S., Soni, V., and Kashaw, S. K. (2024). Drug delivery system approaches for rheumatoid arthritis treatment: a review. Mini-Rev. Med. Chem. 24 (7), 704–720. doi:10.2174/1389557523666230913105803

PubMed Abstract | CrossRef Full Text | Google Scholar

Gong, M., Wen, S., Nguyen, T., Wang, C., Jin, J., and Zhou, L. (2020). Converging relationships of obesity and hyperuricemia with special reference to metabolic disorders and plausible therapeutic implications. Diabetes Metab. Syndr. Obes. 13, 943–962. doi:10.2147/DMSO.S232377

PubMed Abstract | CrossRef Full Text | Google Scholar

Gonzalez, Z., Ferrandez-Montero, A., and Dominguez-Robles, J. (2023). Recent advances in polymers as matrices for drug delivery applications. Pharmaceuticals 16 (12), 1674. doi:10.3390/ph16121674

PubMed Abstract | CrossRef Full Text | Google Scholar

Gulyaev, I. A., Sokol, M. B., Mollaeva, M. R., Klimenko, M. A., Yabbarov, N. G., Chirkina, M. V., et al. (2025). Polymeric drug delivery systems in biomedicine. Biochem.-Moscow. 90 (Suppl. 1), S233–S262. doi:10.1134/S0006297924603976

PubMed Abstract | CrossRef Full Text | Google Scholar

Haddadzadegan, S., Dorkoosh, F., and Bernkop-Schnurch, A. (2022). Oral delivery of therapeutic peptides and proteins: technology landscape of lipid-based nanocarriers. Adv. Drug Deliv. Rev. 182, 114097. doi:10.1016/j.addr.2021.114097

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, T., Wang, M., Li, W., An, M., and Fu, H. (2021). Bmk9 and uricase nanoparticle complex for the treatment of gouty arthritis and uric acid nephropathy. J. Biomed. Nanotechnol. 17 (10), 2071–2084. doi:10.1166/jbn.2021.3168

PubMed Abstract | CrossRef Full Text | Google Scholar

Hao, Y., Li, W., Zhou, X., Yang, F., and Qian, Z. (2017). Microneedles-based transdermal drug delivery systems: a review. J. Biomed. Nanotechnol. 13 (12), 1581–1597. doi:10.1166/jbn.2017.2474

PubMed Abstract | CrossRef Full Text | Google Scholar

Hao, Y., Li, H., Cao, Y., Chen, Y., Lei, M., Zhang, T., et al. (2019). Uricase and horseradish peroxidase hybrid CaHPO₄ nanoflower integrated with transcutaneous patches for treatment of hyperuricemia. J. Biomed. Nanotechnol. 15 (5), 951–965. doi:10.1166/jbn.2019.2752

PubMed Abstract | CrossRef Full Text | Google Scholar

Harun-Or-Rashid, M., Aktar, M. N., Hossain, M. S., Sarkar, N., Islam, M. R., Arafat, M. E., et al. (2023). Recent advances in Micro- and nano-drug delivery systems based on natural and synthetic biomaterials. Polymers 15 (23), 4563. doi:10.3390/polym15234563

PubMed Abstract | CrossRef Full Text | Google Scholar

Harwansh, R. K., Mishra, S., Mazumder, R., Deshmukh, R., and Rahman, A. (2023). Recent updates on transdermal drug delivery approaches for the management of gout and its clinical perspective. Curr. Pharm. Biotechnol. 25, 159–178. doi:10.2174/1389201024666230606143827

PubMed Abstract | CrossRef Full Text | Google Scholar

Hassanzadeh, P., Atyabi, F., and Dinarvand, R. (2019). The significance of artificial intelligence in drug delivery system design. Adv. Drug Deliv. Rev. 151-152, 169–190. doi:10.1016/j.addr.2019.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

He, X., Sun, J., Zhuang, J., Xu, H., Liu, Y., and Wu, D. (2019). Microneedle system for transdermal drug and vaccine delivery: devices, safety, and prospects. Dose-Response 17 (4), 1559325819878585. doi:10.1177/1559325819878585

PubMed Abstract | CrossRef Full Text | Google Scholar

He, J., Tian, W., Meng, Y., Yan, A., Lai, X., Wang, F., et al. (2024). Protective effect of Xylosma congesta extract on renal injury in hyperuricemic rats. Heliyon 10 (23), e40674. doi:10.1016/j.heliyon.2024.e40674

PubMed Abstract | CrossRef Full Text | Google Scholar

Hille, R., Hall, J., and Basu, P. (2014). The mononuclear molybdenum enzymes. Chem. Rev. 114 (7), 3963–4038. doi:10.1021/cr400443z

PubMed Abstract | CrossRef Full Text | Google Scholar

Hooper, M. W., and He, L. (2022). Testing topical products specifically to reduce inflammatory pain from gout: transdermal NSAID delivery and monosodium urate solubility. J. Pain Res. 15, 1825–1835. doi:10.2147/JPR.S367536

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, H., Li, W., Hao, Y., Peng, Z., Zou, Z., Wei, J., et al. (2024). The SGLT2 inhibitor dapagliflozin ameliorates renal fibrosis in hyperuricemic nephropathy. Cell Rep. Med. 5 (8), 101690. doi:10.1016/j.xcrm.2024.101690

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, Q., Lan, H., Tian, Y., Li, X., Wang, M., Zhang, J., et al. (2024). Biofunctional coacervate-based artificial protocells with membrane-like and cytoplasm-like structures for the treatment of persistent hyperuricemia. J. Control. Release. 365, 176–192. doi:10.1016/j.jconrel.2023.11.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Hua, L., Lei, T., Qian, H., Zhang, Y., Hu, Y., and Lei, P. (2021). 3D-printed porous tantalum: recent application in various drug delivery systems to repair hard tissue defects. Expert Opin. Drug Deliv. 18 (5), 625–634. doi:10.1080/17425247.2021.1860015

PubMed Abstract | CrossRef Full Text | Google Scholar

Jafernik, K., Ladniak, A., Blicharska, E., Czarnek, K., Ekiert, H., Wiacek, A. E., et al. (2023). Chitosan-based nanoparticles as effective drug delivery Systems-A review. Molecules 28 (4), 1963. doi:10.3390/molecules28041963

PubMed Abstract | CrossRef Full Text | Google Scholar

Jalal, D. I., Chonchol, M., Chen, W., and Targher, G. (2013). Uric acid as a target of therapy in CKD. Am. J. Kidney. Dis. 61 (1), 134–146. doi:10.1053/j.ajkd.2012.07.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Jampilek, J., and Kralova, K. (2022). Insights into lipid-based delivery nanosystems of protein-tyrosine kinase inhibitors for cancer therapy. Pharmaceutics 14 (12), 2706. doi:10.3390/pharmaceutics14122706

PubMed Abstract | CrossRef Full Text | Google Scholar

Jeong, J., Lee, H., Choi, J., Kang, J., Kim, D., Park, C., et al. (2025). Lung-targeted itraconazole delivery using PVA-based nano-in-microparticles for improved treatment of pulmonary aspergillosis. Int. J. Nanomed. 20, 4357–4380. doi:10.2147/IJN.S511099

PubMed Abstract | CrossRef Full Text | Google Scholar

Jezewski, A. J., Guggisberg, A. M., Hodge, D. M., Ghebremichael, N., John, G. N., Mclellan, L. K., et al. (2022). GAPDH mediates drug resistance and metabolism in Plasmodium falciparum malaria parasites. PLoS Pathog. 18 (9), e1010803. doi:10.1371/journal.ppat.1010803

PubMed Abstract | CrossRef Full Text | Google Scholar

Ji, L., Jiang, W., Huang, J., Xu, Y., Zhou, L., Zhao, Z., et al. (2025). Association of metabolic score for visceral fat (METS-VF) with gout risk in patients with hypertension and hyperuricemia: a multicenter study based on the Chinese population. Diabetes Metab. Syndr. Obes. 18, 761–779. doi:10.2147/DMSO.S502675

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, S., Wang, W., Ke, J., Huang, S., Wang, J., Luo, C., et al. (2023). A mechanically tough and ultra-swellable microneedle for acute gout arthritis. Biomater. Sci. 11 (5), 1714–1724. doi:10.1039/d2bm01937j

PubMed Abstract | CrossRef Full Text | Google Scholar

Ju, S., and Cho, H. (2023). Biohybrid nanoparticle-based in situ monitoring of in vivo drug delivery. Biosensors-Basel. 13 (12), 1017. doi:10.3390/bios13121017

PubMed Abstract | CrossRef Full Text | Google Scholar

Kandav, G., Bhatt, D. C., Jindal, D. K., and Singh, S. K. (2020). Formulation, optimization, and evaluation of allopurinol-loaded bovine serum albumin nanoparticles for targeting kidney in management of hyperuricemic nephrolithiasis: formulation, optimization, and evaluation of ABNPs for kidney targeting. AAPS PharmSciTech 21 (5), 164. doi:10.1208/s12249-020-01695-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Kandav, G., Bhatt, D. C., and Singh, S. K. (2022). Effect of different molecular weights of chitosan on formulation and evaluation of allopurinol-loaded nanoparticles for kidney targeting and in management of hyperuricemic nephrolithiasis. AAPS PharmSciTech 23 (5), 144. doi:10.1208/s12249-022-02297-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, E. H., Shin, A., Park, C. S., Lee, E. B., Lee, Y. J., Curhan, G., et al. (2024). Risk of urolithiasis associated with allopurinol versus benzbromarone among patients with gout: a population-based cohort study. RHEUMATOLOGY 63 (9), 2433–2441. doi:10.1093/rheumatology/keae262

PubMed Abstract | CrossRef Full Text | Google Scholar

Karantas, I. D., Miliotou, A. N., and Siafaka, P. I. (2024). An updated review for hyperuricemia and gout management; special focus on the available drug delivery systems and clinical trials. Curr. Med. Chem. 31 (36), 5856–5883. doi:10.2174/0929867331666230809143758

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaur, U., Pathak, B. K., Meerashahib, T. J., Krishna, D. V. V., and Chakrabarti, S. S. (2024). Should glucokinase be given a chance in diabetes therapeutics? A clinical-pharmacological review of dorzagliatin and lessons learned So far. Clin. Drug Investig. 44 (4), 223–250. doi:10.1007/s40261-024-01351-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Kawachi, Y., Fujishima, Y., Nishizawa, H., Tanaka, A., Yoshida, H., Niwano, S., et al. (2025). Effect of the xanthine oxidase inhibitor febuxostat on the cardio-ankle vascular index in asymptomatic patients with hyperuricemia and liver dysfunction: a sub-analysis of the PRIZE study. J. Atheroscler. Thromb. 32 (4), 474–490. doi:10.5551/jat.65087

PubMed Abstract | CrossRef Full Text | Google Scholar

Kawaguchi, M., Nakayama, A., Aoyagi, Y., Nakamura, T., Shimizu, S., Kawamura, Y., et al. (2021). Both variants of A1CF and BAZ1B genes are associated with gout susceptibility: a replication study and meta-analysis in a Japanese population. Hum. Cell. 34 (2), 293–299. doi:10.1007/s13577-021-00485-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Kemmer, C., Gitzinger, M., Daoud-El Baba, M., Djonov, V., Stelling, J., and Fussenegger, M. (2010). Self-sufficient control of urate homeostasis in mice by a synthetic circuit. Nat. Biotechnol. 28 (4), 355–360. doi:10.1038/nbt.1617

PubMed Abstract | CrossRef Full Text | Google Scholar

Khan, B. A., Naz, F., Alqahtani, A., and Khalid Khan, M. (2024). A nanocomposite competent to overcome solubility and permeation issues of capsaicin and thiocolchicoside simultaneously in gout management: fabrication of nanocubosomes. Saudi Pharm. J. 32 (5), 102050. doi:10.1016/j.jsps.2024.102050

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, S., Kim, M., Jung, S., Kwon, K., Park, J., Kim, S., et al. (2019). Co-delivery of therapeutic protein and catalase-mimic nanoparticle using a biocompatible nanocarrier for enhanced therapeutic effect. J. Control. Release. 309, 181–189. doi:10.1016/j.jconrel.2019.07.038

PubMed Abstract | CrossRef Full Text | Google Scholar

Kivitz, A., Dehaan, W., Azeem, R., Park, J., Rhodes, S., Inshaw, J., et al. (2023). Phase 2 dose-finding study in patients with gout using SEL-212, a novel PEGylated uricase (SEL-037) combined with tolerogenic nanoparticles (SEL-110). Rheumatol. Ther. 10 (4), 825–847. doi:10.1007/s40744-023-00546-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Kluck, V., Liu, R., and Joosten, L. A. B. (2021). The role of interleukin-1 family members in hyperuricemia and gout. Jt. Bone Spine 88 (2), 105092. doi:10.1016/j.jbspin.2020.105092

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, S., and Shukla, R. (2024). Advancements in microneedle technology: current status and next-generation innovations. J. Microencapsul. 41 (8), 782–803. doi:10.1080/02652048.2024.2418613

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, L., and Utreja, P. (2020). Transcending the cutaneous barrier through nanocarrier exploration for passive delivery of anti-hypertensive drugs: a critical review. Nanotechnology 14 (3), 193–209. doi:10.2174/1872210514666200519071734

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, M. S., Kim, S., Lee, J., Bae, Y., and Lee, S. K. (2025). Synovium-on-a-Chip reveals fibroblast-macrophage crosstalk underpinning joint homeostasis and evaluation of gout therapies. Adv. Healthc. Mat. 14, e2501471. doi:10.1002/adhm.202501471

PubMed Abstract | CrossRef Full Text | Google Scholar

Levy, G., and Cheetham, T. C. (2015). Is it time to start treating asymptomatic hyperuricemia? Am. J. Kidney. Dis. 66 (6), 933–935. doi:10.1053/j.ajkd.2015.09.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, D., Dong, J., Xiong, T., Zhou, X., Li, Y., Chen, C., et al. (2024). Transdermal delivery of iguratimod and colchicine ethosome by dissolving microneedle patch for the treatment of recurrent gout. Colloid Surf. B-Biointerfaces 242, 114087. doi:10.1016/j.colsurfb.2024.114087

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, W., Wang, Z., Hao, S., He, H., Wan, Y., Zhu, C., et al. (2017). Mitochondria-targeting polydopamine nanoparticles to deliver doxorubicin for overcoming drug resistance. ACS Appl. Mat. Interfaces. 9 (20), 16793–16802. doi:10.1021/acsami.7b01540

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Q., Liu, P., Wu, C., Bai, L., Zhang, Z., Bao, Z., et al. (2022). Integrating network pharmacology and pharmacological validation to explore the effect of shi wei Ru xiang powder on suppressing hyperuricemia. J. Ethnopharmacol. 298, 115679. doi:10.1016/j.jep.2022.115679

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, D., Yuan, S., Deng, Y., Wang, X., Wu, S., Chen, X., et al. (2023). The dysregulation of immune cells induced by uric acid: mechanisms of inflammation associated with hyperuricemia and its complications. Front. Immunol. 14, 1282890. doi:10.3389/fimmu.2023.1282890

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Z., Xue, L., Yang, J., Wuttke, S., He, P., Lei, C., et al. (2024). Synthetic biohybrids of red blood cells and cascaded-enzymes@ metal-organic frameworks for hyperuricemia treatment. Adv. Sci. 11 (5), e2305126. doi:10.1002/advs.202305126

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, Z., Wu, D., Zhang, H., and Gu, J. (2024). Association between asymptomatic hyperuricemia and risk of arthritis, findings from a US national survey 2007-2018. BMJ Open 14 (2), e074391. doi:10.1136/bmjopen-2023-074391

PubMed Abstract | CrossRef Full Text | Google Scholar

Liao, S., Qiu, G., Hu, Y., Guo, B., and Qiu, Y. (2023). Separable and inseparable silk fibroin microneedles for the transdermal delivery of colchicine: development, characterization, and comparisons. AAPS PharmSciTech 25 (1), 3. doi:10.1208/s12249-023-02716-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Lima, S. A. C., and Reis, S. (2021). Polymeric carriers for biomedical and nanomedicine application. Polymers 13 (8), 1261. doi:10.3390/polym13081261

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Zhu, X., Ji, S., Huang, Z., Zang, Y., Ding, Y., et al. (2022). Transdermal delivery of colchicine using dissolvable microneedle arrays for the treatment of acute gout in a rat model. Drug Deliv. 29 (1), 2984–2994. doi:10.1080/10717544.2022.2122632

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, J., Lin, C., Wu, M., Wang, Y., Chen, S., Yang, T., et al. (2024). Co-delivery of indomethacin and uricase as a new strategy for inflammatory diseases associated with high uric acid. Drug Deliv. Transl. Res. 14 (7), 1820–1838. doi:10.1007/s13346-023-01487-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, G., Liu, J., Shao, P., Kai, D., Yang, L., Sun, P., et al. (2025). Novel nanoliposomes synergistically modulated by sitogluside and dioscin: stability, bioavailability, and capacity to alleviate hyperuricaemia. J. Agric. Food. Chem. 73 (4), 2596–2612. doi:10.1021/acs.jafc.4c08428

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, J., Liu, Z., Li, X., Zhu, L., Xu, G., Chen, Z., et al. (2020). Wireless, battery-free and wearable device for electrically controlled drug delivery: sodium salicylate released from bilayer polypyrrole by near-field communication on smartphone. Biomed. Microdevices 22 (3), 53. doi:10.1007/s10544-020-00511-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, S., Wang, H., Shao, X., Chen, H., Chao, S., Zhang, Y., et al. (2023). Advances in PD-1 signaling inhibition-based nano-delivery systems for tumor therapy. J. Nanobiotechnology. 21 (1), 207. doi:10.1186/s12951-023-01966-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, W., Dai, J., Chen, X., Du, N., and Hu, J. (2025). Integrated network pharmacology and in-silico approaches to decipher the pharmacological mechanism of Dioscorea septemloba thunb in treating gout and its complications. Comb. Chem. High. Throughput Screen. 28 (1), 74–88. doi:10.2174/0113862073258523231025095117

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Zhu, H., Zhou, W., and Ye, Q. (2020). Anti-inflammatory and anti-gouty-arthritic effect of free ginsenoside Rb1 and nano ginsenoside Rb1 against MSU induced gouty arthritis in experimental animals. Chem. Biol. Interact. 332, 109285. doi:10.1016/j.cbi.2020.109285

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Wang, J., and Li, J. (2023). Role of NLRP3 in the pathogenesis and treatment of gout arthritis. Front. Immunol. 14, 1137822. doi:10.3389/fimmu.2023.1137822

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X, X., Song, Z., Tian, W., Huang, Q., Chen, M., Huang, Y., et al. (2025). Advancements in lipid-based delivery systems for functional foods: a comprehensive review of literature and patent trends. Crit. Rev. Food. Sci. Nutr. 65 (13), 2456–2472. doi:10.1080/10408398.2024.2343415

PubMed Abstract | CrossRef Full Text | Google Scholar

Lnu, A., Rathee, G., Sharma, R., Dahiya, S., Vaibhav, F., and Kumar, P. (2024). Treatment adherence and factors influencing it in end-stage renal disease patients on maintenance hemodialysis: a study from a tertiary care hospital in North India. Cureus 16 (11), e73335. doi:10.7759/cureus.73335

PubMed Abstract | CrossRef Full Text | Google Scholar

Losada-Barreiro, S., Celik, S., Sezgin-Bayindir, Z., Bravo-Fernandez, S., and Bravo-Diaz, C. (2024). Carrier systems for advanced drug delivery: improving drug solubility/bioavailability and administration routes. Pharmaceutics 16 (7), 852. doi:10.3390/pharmaceutics16070852

PubMed Abstract | CrossRef Full Text | Google Scholar

Luna, G., Dolzhenko, A. V., and Mancera, R. L. (2025). Synthesis and structure-activity relationship analysis of 2-Substituted-1,2,4-Triazolo[1,5-a]Pyrimidin-7-Ones and their 6-Carboxylate derivatives as xanthine oxidase inhibitors. ChemMedChem. 20 (1), e202400598. doi:10.1002/cmdc.202400598

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, J., Chen, X., Liang, P., Zhao, Z., Wu, T., Li, Z., et al. (2023). Mechanism of anti-hyperuricemia of isobavachin based on network pharmacology and molecular docking. Comput. Biol. Med. 155, 106637. doi:10.1016/j.compbiomed.2023.106637

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, C., Jiang, Y., Xiang, Y., Li, C., Xie, X., Zhang, Y., et al. (2025). Metabolic reprogramming of macrophages by biomimetic melatonin-loaded liposomes effectively attenuates acute gouty arthritis in a mouse model. Adv. Sci. 12 (7), e2410107. doi:10.1002/advs.202410107

PubMed Abstract | CrossRef Full Text | Google Scholar

Mahmoudian, M., Maleki Dizaj, S., Salatin, S., Lobenberg, R., Saadat, M., Islambulchilar, Z., et al. (2021). Oral delivery of solid lipid nanoparticles: underlining the physicochemical characteristics and physiological condition affecting the lipolysis rate. Expert Opin. Drug Deliv. 18 (11), 1707–1722. doi:10.1080/17425247.2021.1982891

PubMed Abstract | CrossRef Full Text | Google Scholar

Manohar, M., Shetty, A., Lobo, C. L., Jain, P., Hebbar, S., Dhas, N., et al. (2025). Emerging trends in chitosan based colloidal drug delivery systems: a translational journey from research to practice. Carbohydr. Polym. 360, 123604. doi:10.1016/j.carbpol.2025.123604

PubMed Abstract | CrossRef Full Text | Google Scholar

Mathew, A. P., Uthaman, S., Cho, K., Cho, C., and Park, I. (2018). Injectable hydrogels for delivering biotherapeutic molecules. Int. J. Biol. Macromol. 110, 17–29. doi:10.1016/j.ijbiomac.2017.11.113

PubMed Abstract | CrossRef Full Text | Google Scholar

Mehmood, A., Althobaiti, F., Zhao, L., Usman, M., Chen, X., Alharthi, F., et al. (2022). Anti-inflammatory potential of stevia residue extract against uric acid-associated renal injury in mice. J. Food Biochem. 46 (10), e14286. doi:10.1111/jfbc.14286

PubMed Abstract | CrossRef Full Text | Google Scholar

Mitnala, S., Phipps-Green, A., Franklin, C., Horne, A., Stamp, L. K., Merriman, T. R., et al. (2016). Clinical and genetic features of diuretic-associated gout: a case-control study. RHEUMATOLOGY 55 (7), 1172–1176. doi:10.1093/rheumatology/kew018

PubMed Abstract | CrossRef Full Text | Google Scholar

Mohamad, S. N. F., Stephanie, S., Sapiun, Z., Tangdilintin, F., Sulistiawati, S., Himawan, A., et al. (2025). Enhanced and sustained transdermal delivery of oxypurinol using thermosensitive gel combined with polymeric solid microneedles. ACS Omega 10 (4), 3500–3510. doi:10.1021/acsomega.4c07716

PubMed Abstract | CrossRef Full Text | Google Scholar

Mohamadi Bian, E., and Moghadam, A. (2025). Synthesis and characterization of Fe(3)O(4)/Chitosan/TiO(2) nanoparticle novel nanocarrier and investigation of in vitro release of vancomycin on HeLa cancer cell. Int. J. Biomater. 2025, 6164871. doi:10.1155/ijbm/6164871

PubMed Abstract | CrossRef Full Text | Google Scholar

Morlock, R. J., Dalal, D., Divino, V., Dekoven, M., Taylor, S. D., Powers, A., et al. (2025). Characteristics and management of uncontrolled gout prior to pegloticase therapy: a 2-year claims analysis. Rheumatol. Ther. 12 (1), 37–51. doi:10.1007/s40744-024-00723-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakayama, A., Kurajoh, M., Toyoda, Y., Takada, T., Ichida, K., and Matsuo, H. (2023). Dysuricemia. Biomedicines 11 (12), 3169. doi:10.3390/biomedicines11123169

PubMed Abstract | CrossRef Full Text | Google Scholar

Narang, R. K., and Dalbeth, N. (2020). Pathophysiology of gout. Semin. Nephrol. 40 (6), 550–563. doi:10.1016/j.semnephrol.2020.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Ng, C. Y., Kee, L. T., Al-Masawa, M. E., Lee, Q. H., Subramaniam, T., Kok, D., et al. (2022). Scalable production of extracellular vesicles and its therapeutic values: a review. Int. J. Mol. Sci. 23 (14), 7986. doi:10.3390/ijms23147986

PubMed Abstract | CrossRef Full Text | Google Scholar

Nguyen, N. N., Mai, T. N. H., Nguyen, B. T., Nguyen, T. D. T., and Tran, T. T. T. (2024). The impact of uric acid-lowering therapy on the progression of non-dialysis chronic kidney disease: a prospective cohort study. Cureus 16 (9), e70435. doi:10.7759/cureus.70435

PubMed Abstract | CrossRef Full Text | Google Scholar

Nishida, Y., Kamatani, N., and Miyamoto, T. (1984). Hypouricaemic effect after oral administration in chickens of polyethylene glycol-modified uricase entrapped in liposomes. J. Pharm. Pharmacol. 36 (5), 354–355. doi:10.1111/j.2042-7158.1984.tb04396.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ohashi, Y., Ooyama, H., Makinoshima, H., Takada, T., Matsuo, H., and Ichida, K. (2024). Plasma and urinary metabolomic analysis of gout and asymptomatic hyperuricemia and profiling of potential biomarkers: a pilot study. Biomedicines 12 (2), 300. doi:10.3390/biomedicines12020300

PubMed Abstract | CrossRef Full Text | Google Scholar

Ozer, I., Kelly, G., Gu, R., Li, X., Zakharov, N., Sirohi, P., et al. (2022). Polyethylene glycol-like brush polymer conjugate of a protein drug does not induce an antipolymer immune response and has enhanced pharmacokinetics than its polyethylene glycol counterpart. Adv. Sci. 9 (11), e2103672. doi:10.1002/advs.202103672

PubMed Abstract | CrossRef Full Text | Google Scholar

Panlu, K., Zhou, Z., Huang, L., Ge, L., Wen, C., and Lv, H. (2024). Associations between obesity and hyperuricemia combing Mendelian randomization with network pharmacology. Heliyon 10 (6), e27074. doi:10.1016/j.heliyon.2024.e27074

PubMed Abstract | CrossRef Full Text | Google Scholar

Parashar, P., Mazhar, I., Kanoujia, J., Yadav, A., Kumar, P., Saraf, S. A., et al. (2022). Appraisal of anti-gout potential of colchicine-loaded chitosan nanoparticle gel in uric acid-induced gout animal model. Arch. Physiol. Biochem. 128 (2), 547–557. doi:10.1080/13813455.2019.1702702

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, K. Y., Kim, H. J., Ahn, H. S., Kim, S. H., Park, E. J., Yim, S., et al. (2016). Effects of coffee consumption on serum uric acid: systematic review and meta-analysis. Semin. Arthritis Rheum. 45 (5), 580–586. doi:10.1016/j.semarthrit.2016.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, S., Chi, S., Yang, J. H., Min, M., and Shin, J. (2024). Comparison of uric acid elevation between aspirin-ticagrelor and aspirin-clopidogrel during dual antiplatelet therapy. Int. J. Clin. Pharmacol. Ther. 62 (11), 534–537. doi:10.5414/CP204606

PubMed Abstract | CrossRef Full Text | Google Scholar

Pascart, T., and Liote, F. (2019). Gout: state of the art after a decade of developments. RHEUMATOLOGY 58 (1), 27–44. doi:10.1093/rheumatology/key002

PubMed Abstract | CrossRef Full Text | Google Scholar

Patel, B., and Thakkar, H. (2023). Formulation development of fast dissolving microneedles loaded with cubosomes of febuxostat: in vitro and in vivo evaluation. Pharmaceutics 15 (1), 224. doi:10.3390/pharmaceutics15010224

PubMed Abstract | CrossRef Full Text | Google Scholar

Paul, B. J., Anoopkumar, K., and Krishnan, V. (2017). Asymptomatic hyperuricemia: is it time to intervene? Clin. Rheumatol. 36 (12), 2637–2644. doi:10.1007/s10067-017-3851-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, X., Li, X., Xie, B., Lai, Y., Sosnik, A., Boucetta, H., et al. (2023). Gout therapeutics and drug delivery. J. Control. Release. 362, 728–754. doi:10.1016/j.jconrel.2023.09.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, Z., Wang, X., Deng, Q., Yan, W., Zhao, W., Pu, Y., et al. (2024). Analysis of dietary vitamin C intake levels and the risk of hyperuricemia and gout based on cross-sectional studies and bi-directional Mendelian randomization. Int. J. Rheum. Dis. 27 (6), e15235. doi:10.1111/1756-185X.15235

CrossRef Full Text | Google Scholar

Pezeshki-Modaress, M., Zandi, M., and Rajabi, S. (2018). Tailoring the gelatin/chitosan electrospun scaffold for application in skin tissue engineering: an in vitro study. Prog. Biomater. 7 (3), 207–218. doi:10.1007/s40204-018-0094-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Piao, M., Wang, H., Jiang, Y., Wu, Y., Nan, J., and Lian, L. (2022). Taxifolin blocks monosodium urate crystal-induced gouty inflammation by regulating phagocytosis and autophagy. Inflammopharmacology 30 (4), 1335–1349. doi:10.1007/s10787-022-01014-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Pino-Zambrano, M. F., Cardoso Penafiel, P., and Calvo-Aranda, E. (2024). Rasburicase plus methotrexate combination therapy in a case of tophaceous, difficult-to-treat gout. Cureus 16 (12), e76265. doi:10.7759/cureus.76265

PubMed Abstract | CrossRef Full Text | Google Scholar

Punzi, L., Scagnellato, L., Galozzi, P., Baggio, C., Damasco, A., Oliviero, F., et al. (2025). Gout: one year in review 2025. Clin. Exp. Rheumatol. 43 (5), 799–808. doi:10.55563/clinexprheumatol/9sdln5

PubMed Abstract | CrossRef Full Text | Google Scholar

Qian, X., Jiang, Y., Luo, Y., and Jiang, Y. (2023). The anti-hyperuricemia and anti-inflammatory effects of Atractylodes macrocephala in hyperuricemia and gouty arthritis rat models. Comb. Chem. High. Throughput Screen. 26 (5), 950–964. doi:10.2174/1386207325666220603101540

PubMed Abstract | CrossRef Full Text | Google Scholar

Quintana, M. J., Shum, A. Z., Folse, M. S., Ramesh, P. C., Ahmadzadeh, S., Varrassi, G., et al. (2023). Gout treatment and clinical considerations: the role of pegloticase, colchicine, and febuxostat. Cureus 15 (10), e46649. doi:10.7759/cureus.46649

PubMed Abstract | CrossRef Full Text | Google Scholar

Rai, A., Shah, K., and Dewangan, H. K. (2023). Review on the artificial intelligence-based nanorobotics targeted drug delivery system for brain-specific targeting. Curr. Pharm. Des. 29 (44), 3519–3531. doi:10.2174/0113816128279248231210172053

PubMed Abstract | CrossRef Full Text | Google Scholar

Rangaraj, N., Shah, S., A J, M., Pailla, S. R., Cheruvu, H. S., D, S., et al. (2019). Quality by design approach for the development of self-emulsifying systems for oral delivery of febuxostat: pharmacokinetic and pharmacodynamic evaluation. AAPS PharmSciTech 20 (7), 267. doi:10.1208/s12249-019-1476-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodriguez, J. M., Garranzo, M., Segura, J., Orgaz, B., Arroyo, R., Alba, C., et al. (2023). A randomized pilot trial assessing the reduction of gout episodes in hyperuricemic patients by oral administration of Ligilactobacillus salivarius CECT 30632, a strain with the ability to degrade purines. Front. Microbiol. 14, 1111652. doi:10.3389/fmicb.2023.1111652

PubMed Abstract | CrossRef Full Text | Google Scholar

Rong, W., Shen, X., Adu-Frimpong, M., He, Q., Zhang, J., Li, X., et al. (2022). Pinocembrin polymeric micellar drug delivery system: preparation, characterisation and anti-hyperuricemic activity evaluation. J. Microencapsul. 39 (5), 419–432. doi:10.1080/02652048.2022.2096138

PubMed Abstract | CrossRef Full Text | Google Scholar

Ruiz-Miyazawa, K. W., Staurengo-Ferrari, L., Pinho-Ribeiro, F. A., Fattori, V., Zaninelli, T. H., Badaro-Garcia, S., et al. (2018). 15d-PGJ(2)-loaded nanocapsules ameliorate experimental gout arthritis by reducing pain and inflammation in a PPAR-gamma-sensitive manner in mice. Sci. Rep. 8 (1), 13979. doi:10.1038/s41598-018-32334-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Salunkhe, S. A., Chitkara, D., Mahato, R. I., and Mittal, A. (2021). Lipid based nanocarriers for effective drug delivery and treatment of diabetes associated liver fibrosis. Adv. Drug Deliv. Rev. 173, 394–415. doi:10.1016/j.addr.2021.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Sands, E., Kivitz, A., Dehaan, W., Leung, S. S., Johnston, L., and Kishimoto, T. K. (2022). Tolerogenic nanoparticles mitigate the formation of anti-drug antibodies against pegylated uricase in patients with hyperuricemia. Nat. Commun. 13 (1), 272. doi:10.1038/s41467-021-27945-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Sattui, S. E., and Gaffo, A. L. (2016). Treatment of hyperuricemia in gout: current therapeutic options, latest developments and clinical implications. Ther. Adv. Musculoskelet. Dis. 8 (4), 145–159. doi:10.1177/1759720X16646703

PubMed Abstract | CrossRef Full Text | Google Scholar

Savla, R., Browne, J., Plassat, V., Wasan, K. M., and Wasan, E. K. (2017). Review and analysis of FDA approved drugs using lipid-based formulations. Drug Dev. Ind. Pharm. 43 (11), 1743–1758. doi:10.1080/03639045.2017.1342654

PubMed Abstract | CrossRef Full Text | Google Scholar

Seo, Y., Lim, H., Park, H., Yu, J., An, J., Yoo, H. Y., et al. (2023). Recent progress of lipid nanoparticles-based lipophilic drug delivery: focus on surface modifications. Pharmaceutics 15 (3), 772. doi:10.3390/pharmaceutics15030772

PubMed Abstract | CrossRef Full Text | Google Scholar

Shao, Y., Chen, C., Yu, X., Yan, J., Guo, J., and Ye, G. (2025). Comprehensive analysis of scRNA-seq and bulk RNA-Seq data via machine learning and bioinformatics reveals the role of lysine metabolism-related genes in gastric carcinogenesis. BMC Cancer 25 (1), 644. doi:10.1186/s12885-025-14051-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, N., Kumar, S., Joshi, G., and Choudhary, D. (2022). Formulation and characterization of febuxostat loaded nanostructured lipid carriers (NLCs) - gel for topical treatment of gout. Nanotechnology 16 (3), 250–258. doi:10.2174/1872210515666210415114118

PubMed Abstract | CrossRef Full Text | Google Scholar

Shin, J. H., Lee, C. M., and Song, J. (2024). Transcutaneous auricular vagus nerve stimulation mitigates gouty inflammation by reducing neutrophil infiltration in BALB/c mice. Sci. Rep. 14 (1), 25630. doi:10.1038/s41598-024-77272-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Silva, A. C., Moreira, J. N., and Lobo, J. M. S. (2022). Editorial-current insights on lipid-based nanosystems. Pharmaceuticals 15 (10), 1267. doi:10.3390/ph15101267

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, H. P., Utreja, P., Tiwary, A. K., and Jain, S. (2009). Elastic liposomal formulation for sustained delivery of colchicine: in vitro characterization and in vivo evaluation of anti-gout activity. AAPS J. 11 (1), 54–64. doi:10.1208/s12248-008-9078-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, N., Parashar, P., Tripathi, C. B., Kanoujia, J., Kaithwas, G., and Saraf, S. A. (2017). Oral delivery of allopurinol niosomes in treatment of gout in animal model. J. Liposome Res. 27 (2), 130–138. doi:10.1080/08982104.2016.1174943

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, J. A., Morlock, A., and Morlock, R. (2025). Gout flare burden in the United States: a multiyear cross-sectional survey study. ACR Open Rheumatol. 7 (1), e11759. doi:10.1002/acr2.11759

PubMed Abstract | CrossRef Full Text | Google Scholar

Sorensen, L. B. (1965). Role of the intestinal tract in the elimination of uric acid. Arthritis Rheum. 8 (5), 694–706. doi:10.1002/art.1780080429

PubMed Abstract | CrossRef Full Text | Google Scholar

Spiga, R., Marini, M. A., Mancuso, E., Di Fatta, C., Fuoco, A., Perticone, F., et al. (2017). Uric acid is associated with inflammatory biomarkers and induces inflammation via activating the NF-κB signaling pathway in HepG2 cells. Arterioscler. Thromb. Vasc. Biol. 37 (6), 1241–1249. doi:10.1161/ATVBAHA.117.309128

PubMed Abstract | CrossRef Full Text | Google Scholar

Stiburkova, B., and Ichida, K. (2025). Genetic background of selected hyperuricemia causing gout with pediatric onset. Jt. Bone Spine 92 (4), 105884. doi:10.1016/j.jbspin.2025.105884

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, J., Liu, X., Du, J., An, J., Li, Y., Hu, Y., et al. (2023). Manganese-doped albumin-gelatin composite nanogel loaded with berberine applied to the treatment of gouty arthritis in rats via a SPARC-Dependent mechanism. Int. J. Biol. Macromol. 253 (Pt 3), 126999. doi:10.1016/j.ijbiomac.2023.126999

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, H., Wang, X., Li, Y., Shen, Y., Zhang, L., Xu, Y., et al. (2024). Targeting liver xor by GalNAc-siRNA is an effective strategy for hyperuricemia therapy. Pharmaceutics 16 (7), 938. doi:10.3390/pharmaceutics16070938

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, W., Cui, L., Terkeltaub, R., Chen, Y., Li, X., Cheng, X., et al. (2025). Risk of hepatotoxicity in patients with gout treated with febuxostat or benzbromarone: a propensity score-matched cohort study. Arthritis Care Res. 77, 1149–1156. doi:10.1002/acr.25547

PubMed Abstract | CrossRef Full Text | Google Scholar

Sundy, J. S., Ganson, N. J., Kelly, S. J., Scarlett, E. L., Rehrig, C. D., Huang, W., et al. (2007). Pharmacokinetics and pharmacodynamics of intravenous PEGylated recombinant Mammalian urate oxidase in patients with refractory gout. Arthritis Rheum. 56 (3), 1021–1028. doi:10.1002/art.22403

PubMed Abstract | CrossRef Full Text | Google Scholar

Tang, Y., Du, Y., Ye, J., Deng, L., and Cui, W. (2024). Intestine-targeted explosive hydrogel microsphere promotes uric acid excretion for gout therapy. Adv. Mat. 36 (3), e2310492. doi:10.1002/adma.202310492

PubMed Abstract | CrossRef Full Text | Google Scholar

Tiwari, S., Dwivedi, H., Kymonil, K. M., and Saraf, S. A. (2015). Urate crystal degradation for treatment of gout: a nanoparticulate combination therapy approach. Drug Deliv. Transl. Res. 5 (3), 219–230. doi:10.1007/s13346-015-0219-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Towiwat, P., Chhana, A., and Dalbeth, N. (2019). The anatomical pathology of gout: a systematic literature review. BMC Musculoskelet. Disord. 20 (1), 140. doi:10.1186/s12891-019-2519-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Tseng, C., Wong, M. C., Liao, W., Chen, C., Lee, S., Yen, J., et al. (2020). Systemic investigation of promoter-wide methylome and genome variations in gout. Int. J. Mol. Sci. 21 (13), 4702. doi:10.3390/ijms21134702

PubMed Abstract | CrossRef Full Text | Google Scholar

Tuono, R. D. M., Samou, A. B. F., Mbiandjeu, M. T., Well A Well, P. B. A. K., and Chuisseu, P. D. D. (2024). Hyperuricemia and associated factors: the case of outpatients at the bafoussam regional Hospital- Cameroon, an analytical cross-sectional study. Health Sci. Rep. 7 (2), e1891. doi:10.1002/hsr2.1891

PubMed Abstract | CrossRef Full Text | Google Scholar

Uskudar Cansu, D., Erdogan, B., and Korkmaz, C. (2019). Can hyperuricemia predict glycogen storage disease (mcArdle's disease) in rheumatology practice? (myogenic hyperuricemia). Clin. Rheumatol. 38 (10), 2941–2948. doi:10.1007/s10067-019-04572-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Valsalan Soba, S., Babu, M., and Panonnummal, R. (2021). Ethosomal gel formulation of alpha phellandrene for the transdermal delivery in gout. Adv. Pharm. Bull. 11 (1), 137–149. doi:10.34172/apb.2021.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Valsaraj, R., Singh, A. K., Gangopadhyay, K. K., Ghoshdastidar, B., Goyal, G., Batin, M., et al. (2020). Management of asymptomatic hyperuricemia: Integrated diabetes and endocrine academy (IDEA) consensus statement. Diabetes Metab. syndr.-clin. Res. Rev. 14 (2), 93–100. doi:10.1016/j.dsx.2020.01.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Waheed, Y., Yang, F., and Sun, D. (2021). Role of asymptomatic hyperuricemia in the progression of chronic kidney disease and cardiovascular disease. J. Intern. Med. 36 (6), 1281–1293. doi:10.3904/kjim.2020.340

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Luo, C., Yang, G., Wei, X., Liu, D., and Zhou, S. (2016). A luteolin-loaded electrospun fibrous implantable device for potential therapy of gout attacks. Macromol. Biosci. 16 (11), 1598–1609. doi:10.1002/mabi.201600123

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, R., Wang, H., Jiang, G., Sun, Y., Liu, T., Nie, L., et al. (2023). Transdermal delivery of allopurinol to acute hyperuricemic mice via polymer microneedles for the regulation of serum uric acid levels. Biomater. Sci. 11 (5), 1704–1713. doi:10.1039/d2bm01836e

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Wang, Z., Jiang, H., Ni, L., Ju, H., Wu, Y., et al. (2025). Advances in the use of nanotechnology for treating gout. Nanomedicine 20 (4), 355–369. doi:10.1080/17435889.2025.2457315

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, H., Zhang, H., Sun, L., and Guo, W. (2018). Roles of hyperuricemia in metabolic syndrome and cardiac-kidney-vascular system diseases. Am. J. Transl. Res. 10 (9), 2749–2763.

PubMed Abstract | Google Scholar

Wang, Q., Yang, Q., Cao, X., Wei, Q., Firempong, C. K., Guo, M., et al. (2018). Enhanced oral bioavailability and anti-gout activity of [6]-shogaol-loaded solid lipid nanoparticles. Int. J. Pharm. 550 (1-2), 24–34. doi:10.1016/j.ijpharm.2018.08.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, R., Sun, Y., Wang, H., Liu, T., Shavandi, A., Nie, L., et al. (2024). Core-shell structured microneedles with programmed drug release functions for prolonged hyperuricemia management. J. Mat. Chem. B 12 (4), 1064–1076. doi:10.1039/d3tb02607h

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, X., Zhao, J., Lin, Z., Li, J., Li, X., Xu, X., et al. (2024). Analysis of polyphenol extract from hazel leaf and ameliorative efficacy and mechanism against hyperuricemia zebrafish model via network pharmacology and molecular docking. Molecules 29 (2), 317. doi:10.3390/molecules29020317

PubMed Abstract | CrossRef Full Text | Google Scholar

Weaver, D. J. J. (2019). Uric acid and progression of chronic kidney disease. Pediatr. Nephrol. 34 (5), 801–809. doi:10.1007/s00467-018-3979-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, C., Wang, Q., Weng, W., Adu-Frimpong, M., Toreniyazov, E., Ji, H., et al. (2022). Enhanced oral bioavailability and anti-hyperuricemic activity of liquiritin via a self-nanoemulsifying drug delivery system. J. Sci. Food. Agric. 102 (5), 2032–2040. doi:10.1002/jsfa.11542

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, Y., Zhang, Y., Lu, X., Zhou, J., Cheng, H., Liu, Y., et al. (2025). Novel colchicine ethosomes cataplasm for the treatment of acute gouty arthritis. Colloid Surf. B-Biointerfaces 254, 114776. doi:10.1016/j.colsurfb.2025.114776

PubMed Abstract | CrossRef Full Text | Google Scholar

Wen, S., Arakawa, H., and Tamai, I. (2024). Uric acid in health and disease: from physiological functions to pathogenic mechanisms. Pharmacol. Ther. 256, 108615. doi:10.1016/j.pharmthera.2024.108615

PubMed Abstract | CrossRef Full Text | Google Scholar

Weng, W., Wang, Q., Wei, C., Man, N., Zhang, K., Wei, Q., et al. (2019). Preparation, characterization, pharmacokinetics and anti-hyperuricemia activity studies of myricitrin-loaded proliposomes. Int. J. Pharm. 572, 118735. doi:10.1016/j.ijpharm.2019.118735

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X., and You, C. (2023). The biomarkers discovery of hyperuricemia and gout: proteomics and metabolomics. PeerJ 11, e14554. doi:10.7717/peerj.14554

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, D., Yang, S., Yuan, C., Zhang, K., Tan, J., Guan, K., et al. (2024). Targeting purine metabolism-related enzymes for therapeutic intervention: a review from molecular mechanism to therapeutic breakthrough. Int. J. Biol. Macromol. 282 (Pt 1), 136828. doi:10.1016/j.ijbiomac.2024.136828

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y., Li, P., Jiang, Z., Sun, X., He, H., Yan, P., et al. (2023). Bioinspired yeast-based beta-glucan system for oral drug delivery. Carbohydr. Polym. 319, 121163. doi:10.1016/j.carbpol.2023.121163

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, F., Chen, L., and Du, Y. (2024). Comparison of the efficacy and safety of benzbromarone and febuxostat in gout and hyperuricemia: a systematic review and meta-analysis. Clin. Rheumatol. 43 (5), 1745–1754. doi:10.1007/s10067-024-06933-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiang, G., Yang, L., Qin, J., Wang, S., Zhang, Y., and Yang, S. (2024). Revealing the potential bioactive components and mechanism of qianhua gout capsules in the treatment of gouty arthritis through network pharmacology, molecular docking and pharmacodynamic study strategies. Heliyon 10 (10), e30983. doi:10.1016/j.heliyon.2024.e30983

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiong, H., Zhou, Y., Zhou, Q., He, D., Deng, X., Sun, Q., et al. (2016). Nanocapsule assemblies as effective enzyme delivery systems against hyperuricemia. Nanomedicine 12 (6), 1557–1566. doi:10.1016/j.nano.2016.02.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, C., Wan, X., Xu, L., Weng, H., Yan, M., Miao, M., et al. (2015). Xanthine oxidase in non-alcoholic fatty liver disease and hyperuricemia: one stone hits two birds. J. Hepatol. 62 (6), 1412–1419. doi:10.1016/j.jhep.2015.01.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, J., Wu, M., Yang, J., Zhao, D., He, D., Liu, Y., et al. (2024). Multimodal smart systems reprogramme macrophages and remove urate to treat gouty arthritis. Nat. Nanotechnol. 19 (10), 1544–1557. doi:10.1038/s41565-024-01715-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, J., Yu, X., Li, Q., Miao, M., and Shao, Y. (2024). Machine learning to establish three sphingolipid metabolism genes signature to characterize the immune landscape and prognosis of patients with gastric cancer. BMC Genomics 25 (1), 319. doi:10.1186/s12864-024-10243-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, W., Wang, X., Peng, Z., Deng, Q., Zhu, M., Meng, F., et al. (2025). A bidirectional mendelian randomization study of tea-drinking habits and risk of elevated serum uric acid levels. Int. J. Rheum. Dis. 28 (2), e70128. doi:10.1111/1756-185X.70128

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, B., Xin, M., Liang, S., Huang, Y., Li, J., Wang, C., et al. (2023). Naringenin ameliorates hyperuricemia by regulating renal uric acid excretion via the PI3K/AKT signaling pathway and renal inflammation through the NF-κB signaling pathway. J. Agric. Food. Chem. 71 (3), 1434–1446. doi:10.1021/acs.jafc.2c01513

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, L., Zhang, Y., Xie, J., Zhong, C., He, D., Wang, T., et al. (2020). Biomimetic polysaccharide-cloaked lipidic nanovesicles/microassemblies for improving the enzymatic activity and prolonging the action time for hyperuricemia treatment. Nanoscale 12 (28), 15222–15235. doi:10.1039/d0nr02651d

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, S., Liu, H., Fang, X., Yan, F., and Zhang, Y. (2024). Signaling pathways in uric acid homeostasis and gout: from pathogenesis to therapeutic interventions. Int. Immunopharmacol. 132, 111932. doi:10.1016/j.intimp.2024.111932

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, M., Gu, Y., Tang, X., Wang, T., and Liu, J. (2019). Advancement of lipid-based nanocarriers and combination application with physical penetration technique. Curr. Drug Deliv. 16 (4), 312–324. doi:10.2174/1567201816666190118125427

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Q., Wang, Q., Feng, Y., Wei, Q., Sun, C., Firempong, C. K., et al. (2019). Anti-hyperuricemic property of 6-shogaol via self-micro emulsifying drug delivery system in model rats: formulation design, in vitro and in vivo evaluation. Drug Dev. Ind. Pharm. 45 (8), 1265–1276. doi:10.1080/03639045.2019.1594885

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Y., Li, Z., Huang, P., Lin, J., Li, J., Shi, K., et al. (2023). Rapidly separating dissolving microneedles with sustained-release colchicine and stabilized uricase for simplified long-term gout management. Acta Pharm. Sin. B 13 (8), 3454–3470. doi:10.1016/j.apsb.2023.02.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Yao, T., Lee, R., Wu, W., Chen, I., Yu, T., and Yeh, K. (2024). Advances in gouty arthritis management: integration of established therapies, emerging treatments, and lifestyle interventions. Int. J. Mol. Sci. 25 (19), 10853. doi:10.3390/ijms251910853

PubMed Abstract | CrossRef Full Text | Google Scholar

Yi, H., Yu, H., Wang, L., Wang, Y., Ouyang, C., and Keshta, B. E. (2024). Microneedle transdermal drug delivery as a candidate for the treatment of gouty arthritis: material structure, design strategies and prospects. Acta Biomater. 187, 20–50. doi:10.1016/j.actbio.2024.08.032

PubMed Abstract | CrossRef Full Text | Google Scholar

You, B., and Chen, C. (2024). Fluidic membrane-bound protocells enabling versatile assembly of functional nanomaterials for biomedical applications. ACS Nano 18 (49), 33545–33554. doi:10.1021/acsnano.4c11349

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, Z., Wang, H., and You, G. (2023). The regulation of human organic anion transporter 4 by insulin-like growth factor 1 and protein kinase B signaling. Biochem. Pharmacol. 215, 115702. doi:10.1016/j.bcp.2023.115702

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng, Y., Li, S., Liu, C., Gong, T., Sun, X., Fu, Y., et al. (2017). Soluplus micelles for improving the oral bioavailability of scopoletin and their hypouricemic effect in vivo. Acta Pharmacol. Sin. 38 (3), 424–433. doi:10.1038/aps.2016.126

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng, Y., Ma, Y., Yang, Z., Mao, J., and Zheng, Y. (2020). Antihyperuricemic efficacy of Scopoletin-loaded soluplus micelles in yeast extract/potassium oxonate-induced hyperuricemic mice. Drug Dev. Ind. Pharm. 46 (9), 1550–1557. doi:10.1080/03639045.2020.1811302

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng, L., Deng, Y., Zhou, X., Ji, S., Peng, B., Lu, H., et al. (2024). Simiao pills alleviates renal injury associated with hyperuricemia: a multi-omics analysis. J. Ethnopharmacol. 333, 118492. doi:10.1016/j.jep.2024.118492

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, W. (2023). Uric acid en route to gout. Adv. Clin. Chem. 116, 209–275. doi:10.1016/bs.acc.2023.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Neogi, T., Chen, C., Chaisson, C., Hunter, D. J., and Choi, H. K. (2012). Cherry consumption and decreased risk of recurrent gout attacks. Arthritis Rheum. 64 (12), 4004–4011. doi:10.1002/art.34677

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, J., Shuai, X., Li, J., Xiang, N., Gong, T., and Zhang, Z. (2016). Biodistribution, hypouricemic efficacy and therapeutic mechanism of morin phospholipid complex loaded self-nanoemulsifying drug delivery systems in an experimental hyperuricemic model in rats. J. Pharm. Pharmacol. 68 (1), 14–25. doi:10.1111/jphp.12492

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X., Xu, D., Jin, X., Liu, G., Liang, S., Wang, H., et al. (2017). Nanocapsules of therapeutic proteins with enhanced stability and long blood circulation for hyperuricemia management. J. Control. Release. 255, 54–61. doi:10.1016/j.jconrel.2017.03.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, J., Sun, Y., Qu, Q., Li, B., Zhang, L., Gu, R., et al. (2021). Engineering non-covalently assembled protein nanoparticles for long-acting gouty arthritis therapy. J. Mat. Chem. B 9 (48), 9923–9931. doi:10.1039/d1tb01760h

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, F., Gan, Y., Xie, W., Lu, S., Zha, Y., Liang, Y., et al. (2025). A novel zinc ferrite nanoparticle protects against MSU-Induced gout arthritis via Nrf2/NF-κB/NLRP3 pathway. Life Sci. 366-367, 123475. doi:10.1016/j.lfs.2025.123475

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, K., Wang, Q., Yang, Q., Wei, Q., Man, N., Adu-Frimpong, M., et al. (2019). Enhancement of oral bioavailability and anti-hyperuricemic activity of isoliquiritigenin via self-microemulsifying drug delivery system. AAPS PharmSciTech 20 (5), 218. doi:10.1208/s12249-019-1421-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, S., Wang, Y., Cheng, J., Huangfu, N., Zhao, R., Xu, Z., et al. (2019). Hyperuricemia and cardiovascular disease. Curr. Pharm. Des. 25 (6), 700–709. doi:10.2174/1381612825666190408122557

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Zhang, N., Song, H., Li, H., Wen, J., Tan, X., et al. (2019). Design, characterization and comparison of transdermal delivery of colchicine via borneol-chemically-modified and borneol-physically-modified ethosome. Drug Deliv. 26 (1), 70–77. doi:10.1080/10717544.2018.1559258

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, C., and Zhou, B. (2022). Polyethyleneimine-based drug delivery systems for cancer theranostics. J. Funct. Biomater. 14 (1), 12. doi:10.3390/jfb14010012

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, Z., Chen, Y., and Shi, Y. (2020). Microneedles: a potential strategy in transdermal delivery and application in the management of psoriasis. RSC Adv. 10 (24), 14040–14049. doi:10.1039/d0ra00735h

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, J., Guo, S., Schrodi, S. J., and He, D. (2022). Trends in the contribution of genetic susceptibility loci to hyperuricemia and gout and associated novel mechanisms. Front. Cell. Dev. Biol. 10, 937855. doi:10.3389/fcell.2022.937855

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, P., Hu, H., Chen, X., Jiang, Q., Yu, X., Cen, X., et al. (2024). Mild hyperthermia enhanced synergistic uric acid degradation and multiple ROS elimination for an effective acute gout therapy. J. Nanobiotechnology. 22 (1), 275. doi:10.1186/s12951-024-02539-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, L., Chang, R., Liang, B., Wang, Y., Zhu, Y., Jia, Z., et al. (2024). Overcoming drug resistance through extracellular vesicle-based drug delivery system in cancer treatment. Cancer Drug Resist 7, 50. doi:10.20517/cdr.2024.107

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhong, Y., Yang, C., Zhang, B., Chen, Y., Cai, W., Wang, G., et al. (2024). Causal impact of human blood metabolites and metabolic pathways on serum uric acid and gout: a Mendelian randomization study. Front. Endocrinol. 15, 1378645. doi:10.3389/fendo.2024.1378645

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, Z., Liu, J., Yang, Y., Adu-Frimpong, M., Ji, H., Toreniyazov, E., et al. (2021). SMEDDS for improved oral bioavailability and anti-hyperuricemic activity of licochalcone A. J. Microencapsul. 38 (7-8), 459–471. doi:10.1080/02652048.2021.1963341

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, M., Wang, X., Peng, Z., Yan, W., Deng, Q., Li, M., et al. (2025). The role of the estimated glomerular filtration rate and body roundness index in the risk assessment of uric acid-lowering therapy-resistant gout in U.S. adults: evidence from the national health and nutrition examination survey (2007-2018). Ren. Fail. 47 (1), 2441398. doi:10.1080/0886022X.2024.2441398

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: gout, hyperuricemia, drug delivery systems, therapeutic, prospect

Citation: Peng Z, Meng F, Deng Q, Huang Y, Mao D, Long Y, Yan W, Peng J, Wang X and Liu N (2025) Advances in drug delivery systems for the management of gout and hyperuricemia. Front. Pharmacol. 16:1660890. doi: 10.3389/fphar.2025.1660890

Received: 08 July 2025; Accepted: 04 September 2025;
Published: 01 October 2025.

Edited by:

Sarmistha Saha, GLA University, India

Reviewed by:

Sidharth Mehan, Indo-Soviet Friendship College of Pharmacy, India
Pratibha Singh, All India Institute of Medical Sciences, India

Copyright © 2025 Peng, Meng, Deng, Huang, Mao, Long, Yan, Peng, Wang and Liu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jiangyun Peng, cGVuZ2ppYW5neXVuQDEyNi5jb20=; Xingqiang Wang, b3V5YW5nZmVuZ3lhbmdndW9AMTYzLmNvbQ==; Nian Liu, bGl1bmlhbjg5MTdAMTYzLmNvbQ==

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.