Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 357
1.
Nano Lett ; 24(22): 6634-6643, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38742828

The effect of strong metal-support interaction (SMSI) has never been systematically studied in the field of nanozyme-based catalysis before. Herein, by coupling two different Pd crystal facets with MnO2, i.e., (100) by Pd cube (Pdc) and (111) by Pd icosahedron (Pdi), we observed the reconstruction of Pd atomic structure within the Pd-MnO2 interface, with the reconstructed Pdc (100) facet more disordered than Pdi (111), verifying the existence of SMSI in such coupled system. The rearranged Pd atoms in the interface resulted in enhanced uricase-like catalytic activity, with Pdc@MnO2 demonstrating the best catalytic performance. Theoretical calculations suggested that a more disordered Pd interface led to stronger interactions with intermediates during the uricolytic process. In vitro cell experiments and in vivo therapy results demonstrated excellent biocompatibility, therapeutic effect, and biosafety for their potential hyperuricemia treatment. Our work provides a brand-new perspective for the design of highly efficient uricase-mimic catalysts.


Hyperuricemia , Manganese Compounds , Oxides , Urate Oxidase , Hyperuricemia/drug therapy , Urate Oxidase/chemistry , Urate Oxidase/therapeutic use , Urate Oxidase/metabolism , Oxides/chemistry , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Humans , Palladium/chemistry , Palladium/pharmacology , Animals , Catalysis , Uric Acid/chemistry , Mice
2.
Int J Hematol ; 119(6): 660-666, 2024 Jun.
Article En | MEDLINE | ID: mdl-38575822

Guidelines recommend rasburicase for high-risk patients to prevent tumor lysis syndrome (TLS). However, little information is available on the incidence and outcome of TLS in AML patients. We analyzed 145 patients with AML who underwent induction therapy before the approval of rasburicase to evaluate the incidence of TLS and the necessity of rasburicase as prophylaxis. Three patients had already developed clinical TLS (CTLS) at diagnosis of AML, and another three developed CTLS after the initiation of chemotherapy. In patients without TLS at diagnosis of AML, the risk for developing TLS was classified as high in 44 patients, intermediate in 41 and low in 57, according to the current guidelines. Allopurinol alone was administered to prevent hyperuricemia in all patients. All three patients who developed CTLS after diagnosis of AML were at high risk of TLS, and had elevated serum creatinine levels and a WBC count greater than 200,000 per microliter at diagnosis of AML. Allopurinol may be insufficient to prevent TLS in high-risk patients with renal dysfunction at diagnosis of AML, especially those with a high tumor burden and a WBC count of 200,000 or more, which indicates that prophylactic administration of rasburicase should be considered.


Allopurinol , Leukemia, Myeloid, Acute , Tumor Lysis Syndrome , Urate Oxidase , Humans , Tumor Lysis Syndrome/etiology , Tumor Lysis Syndrome/prevention & control , Urate Oxidase/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/complications , Male , Female , Middle Aged , Allopurinol/therapeutic use , Allopurinol/administration & dosage , Aged , Adult , Induction Chemotherapy , Aged, 80 and over , Hyperuricemia/drug therapy , Adolescent , Incidence , Young Adult
3.
Int J Mol Sci ; 25(6)2024 Mar 15.
Article En | MEDLINE | ID: mdl-38542302

Cardiorenal syndrome (CRS) involves joint dysfunction of the heart and kidney. Acute forms share biochemical alterations like hyperuricaemia (HU) with tumour lysis syndrome (TLS). The mainstay treatment of acute CRS with systemic overload is diuretics, but rasburicase is used in TLS to prevent and treat hyperuricaemia. An observational, retrospective study was performed to assess the effectiveness and safety of a single dose of rasburicase in hospitalized patients with cardiorenal syndrome, worsening renal function and uric acid levels above 9 mg/dL. Rasburicase improved diuresis and systemic congestion in the 35 patients included. A total of 86% of patients did not need to undergo RRT, and early withdrawal was possible in the remaining five. Creatinine (Cr) decreased after treatment with rasburicase from a peak of 3.6 ± 1.27 to 1.79 ± 0.83 mg/dL, and the estimated glomerular filtration rate (eGFR) improved from 17 ± 8 to 41 ± 20 mL/min/1.73 m2 (p = 0.0001). The levels of N-terminal type B Brain Natriuretic Peptide (Nt-ProBNP) and C-reactive protein (CRP) were also significantly reduced. No relevant adverse events were detected. Our results show that early treatment with a dose of rasburicase in patients with CRS and severe HU is effective to improve renal function and systemic congestion, avoiding the need for sustained extrarenal clearance, regardless of comorbidities and ventricular function.


Cardio-Renal Syndrome , Hyperuricemia , Tumor Lysis Syndrome , Humans , Hyperuricemia/drug therapy , Cardio-Renal Syndrome/drug therapy , Retrospective Studies , Tumor Lysis Syndrome/drug therapy , Tumor Lysis Syndrome/etiology , Tumor Lysis Syndrome/prevention & control , Urate Oxidase/therapeutic use
4.
Nano Lett ; 24(11): 3432-3440, 2024 Mar 20.
Article En | MEDLINE | ID: mdl-38391135

Uricase-catalyzed uric acid (UA) degradation has been applied for hyperuricemia therapy, but this medication is limited by H2O2 accumulation, which can cause oxidative stress of cells, resulting in many other health issues. Herein, we report a robust cubic hollow nanocage (HNC) system based on polyvinylpyrrolidone-coated PdPt3 and PdIr3 to serve as highly efficient self-cascade uricase/peroxidase mimics to achieve the desired dual catalysis for both UA degradation and H2O2 elimination. These HNCs have hollow cubic shape with average wall thickness of 1.5 nm, providing desired synergy to enhance catalyst's activity and stability. Density functional theory calculations suggest the PdIr3 HNC surface tend to promote OH*/O* desorption for better peroxidase-like catalysis, while the PdPt3 HNC surface accelerates the UA oxidation by facilitating O2-to-H2O2 conversion. The dual catalysis power demonstrated by these HNCs in cell studies suggests their great potential as a new type of nanozyme for treating hyperuricemia.


Hyperuricemia , Peroxidase , Humans , Peroxidase/therapeutic use , Urate Oxidase/therapeutic use , Povidone/therapeutic use , Hyperuricemia/drug therapy , Hydrogen Peroxide , Uric Acid/metabolism , Oxidoreductases , Coloring Agents
5.
Rheumatology (Oxford) ; 63(4): 1058-1067, 2024 Apr 02.
Article En | MEDLINE | ID: mdl-37449908

OBJECTIVES: Serum urate (SU) lowering with PEGylated uricases in gout can reduce flares and tophi. However, treatment-emergent anti-drug antibodies adversely affect safety and efficacy and the currently approved PEGylated uricase pegloticase requires twice-monthly infusions. Investigational SEL-212 therapy aims to promote uricase-specific tolerance via monthly sequential infusions of a proprietary rapamycin-containing nanoparticle (ImmTOR) and pegadricase. METHODS: COMPARE was a randomized, phase 2, open-label trial of SEL-212 vs pegloticase in adults with refractory gout. SEL-212 [ImmTOR (0.15 mg/kg) and pegadricase (0.2 mg/kg)] was infused monthly or pegloticase (8 mg) twice monthly for 6 months. The primary endpoint was the proportion of participants with SU <6 mg/dl for ≥80% of the time during 3 and 6 months. Secondary outcomes were mean SU, gout flares, number of tender and/or swollen joints and safety. RESULTS: During months 3 and 6 combined, numerically more participants achieved and maintained a SU <6 mg/dl for ≥80% of the time with SEL-212 vs pegloticase (53.0% vs 46.0%, P = 0.181). The percentage reductions in SU levels were statistically greater during months 3 and 6 with SEL-212 vs pegloticase (-73.79% and -47.96%, P = 0.0161). Reductions in gout flare incidence and number of tender and/or swollen joints were comparable between treatments. There were numerical differences between the most common treatment-related adverse events of interest with SEL-212 and pegloticase: gout flares (60.2% vs 50.6%), infections (25.3% vs 18.4%) and infusion-related reactions (15.7% vs 11.5%), respectively. Stomatitis (and related terms) was experienced by eight participants (9.6%) with SEL-212 and none with pegloticase. Stomatitis, a known event for rapamycin, was associated with ImmTOR only. CONCLUSIONS: SEL-212 efficacy and tolerability were comparable to pegloticase in refractory gout. This was associated with a substantial reduction in treatment burden with SEL-212 due to decreased infusion frequency vs pegloticase. CLINICAL TRIAL REGISTRATION: NCT03905512.


Gout , Stomatitis , Adult , Humans , Urate Oxidase/therapeutic use , Urate Oxidase/adverse effects , Gout Suppressants/adverse effects , Uric Acid , Treatment Outcome , Symptom Flare Up , Polyethylene Glycols/adverse effects , Uricosuric Agents/therapeutic use , Stomatitis/chemically induced , Stomatitis/drug therapy
6.
Drug Deliv Transl Res ; 14(7): 1820-1838, 2024 Jul.
Article En | MEDLINE | ID: mdl-38127247

Uric acid is the final metabolite in humans. High level of uric acid chronically induces urate deposition, aggravates kidney damage, and concomitantly causes an increase in inflammatory factors. Alleviating acute inflammation and decreasing uric acid levels are the key points in the treatment of inflammatory diseases associated with high uric acid. However, a drug delivery system that combines anti-inflammatory and uric acid reduction functions at the same time remains a challenge to be settled. Here, we designed a nanocrystal-based co-delivery platform, IND Nplex, characterized by loading of indomethacin (IND) and uricase. Compared with free IND or uricase, IND Nplex possessed a better anti-inflammatory effect by restraining the release of inflammation-related factors in vitro. In addition, pharmacokinetic and biodistribution studies revealed that IND Nplex significantly prolonged the retention time in vivo and was more concentrated in the kidney. In acute gouty arthritis model rats, IND Nplex markedly relieved ankle joint swelling and mitigated synovial inflammation. In acute kidney injury model rats, IND Nplex indicated better biocompatibility and significant amelioration of renal fibrosis. Moreover, IND Nplex showed the effect of anti-inflammatory and improved renal function via determination of inflammatory factors and biochemical markers in the serum and kidney. In conclusion, these results indicate that IND Nplex exerts anti-inflammatory activity and uric acid-lowering effect and could become a promising candidate for the treatment of uric acid-related diseases.


Arthritis, Gouty , Indomethacin , Rats, Sprague-Dawley , Urate Oxidase , Uric Acid , Indomethacin/administration & dosage , Animals , Urate Oxidase/administration & dosage , Urate Oxidase/pharmacokinetics , Urate Oxidase/therapeutic use , Uric Acid/blood , Male , Arthritis, Gouty/drug therapy , Nanoparticles/administration & dosage , Rats , Mice , Inflammation/drug therapy , Tissue Distribution , Acute Kidney Injury/drug therapy , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Drug Delivery Systems , Kidney/drug effects , Kidney/metabolism , Humans , RAW 264.7 Cells , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/pharmacology
7.
Curr Rheumatol Rep ; 26(3): 69-80, 2024 03.
Article En | MEDLINE | ID: mdl-38133712

PURPOSE OF REVIEW: Despite effective available treatments, gout management is often unsuccessful in getting patients to target serum urate goal and in managing flares in the setting of comorbidities. Studies addressing future treatment options for short- and long-term management are reviewed. RECENT FINDINGS: URAT-1 blocking agents have been helpful but have had limitations related to effects on renal function, lack of efficacy with renal impairment, and potential to increase renal stones. Dotinurad may function in the setting of decreased renal function. Arhalofenate has anti-URAT-1 activity and may also blunt gout flares. A new xanthine oxidase inhibitor (XOI), tigulixostat, is under study. New uricase treatments manufactured in combination with agents that can reduce immunogenicity may make uricase treatment simpler. A unique strategy of inhibiting gut uricase may offer the benefits of avoiding systemic absorption. For gout flares, IL-1ß inhibitor studies in progress include different dosing schedules. Dapansutrile, an oral agent under investigation, inhibits activation of the NLRP3 inflammasome and may be an effective anti-inflammatory. New treatments for gout that are under study may work in the setting of comorbidities, simplify management, utilize new mechanisms, or have reduced side effects.


Gout , Hyperuricemia , Humans , Gout Suppressants/therapeutic use , Urate Oxidase/therapeutic use , Uric Acid , Gout/drug therapy , Anti-Inflammatory Agents/therapeutic use , Hyperuricemia/drug therapy
8.
Arthritis Res Ther ; 25(1): 247, 2023 12 18.
Article En | MEDLINE | ID: mdl-38111075

BACKGROUND: Exogenously providing engineered Uox with enhanced half-life is one of the important urate-lowering treatments for gout. The potential of PAT101, a recombinant human albumin (rHA)-conjugated variant, was evaluated and compared as a novel gout treatment through various in vivo studies with PAT101 and competing drugs. METHODS: PAT101 was produced by site-specific conjugation of rHA and Aspergillus flavus Uox (AfUox-rHA) through clickable non-natural amino acid (frTet) and Inverse electron demand Diels-Alder (IEDDA) reaction. In vivo pharmacokinetics, efficacy tests and in vitro immunogenetic assay were performed after single or multiple doses of PAT101 and its competitors in BALB/c mice, transgenic (TG) mice, Sprague-Dawley (SD) rats, and non-human primate (NHP). RESULTS: The half-life of PAT101 in single-dose treated TG mice was more than doubled compared to pegloticase. In SD rats with 4 weeks of repeated administration of rasburicase, only 24% of Uox activity remained, whereas in PAT101, it was maintained by 86%. In the Uox KO model, the survival rate of PAT101 was comparable to that of pegloticase. In addition, human PBMC-based CD4+/CD8+ T-cell activation analysis demonstrated that PAT101 has a lower immune response compared to the original drug, rasburicase. CONCLUSION: All results suggest that this rHA-conjugated AfUox, PAT101, can be provided as a reliable source of Uox for gout treatment.


Gout , Urate Oxidase , Mice , Animals , Rats , Humans , Urate Oxidase/therapeutic use , Leukocytes, Mononuclear/metabolism , Rats, Sprague-Dawley , Gout/drug therapy , Gout Suppressants/therapeutic use , Mice, Transgenic , Polyethylene Glycols/therapeutic use , Albumins/therapeutic use
9.
Acta Biomater ; 170: 250-259, 2023 10 15.
Article En | MEDLINE | ID: mdl-37659730

The interactions between polymers and the immune system remains poorly controlled. In some instances, the immune system can produce antibodies specific to polymer constituents. Indeed, roughly half of pegloticase patients without immunomodulation develop high titers of anti-PEG antibodies (APA) to the PEG polymers on pegloticase, which then quickly clear the drug from circulation and render the gout treatment ineffective. Here, using pegloticase as a model drug, we show that addition of high molecular weight (MW) free (unconjugated) PEG to pegloticase allows us to control the immunogenicity and mitigates APA induction in mice. Compared to pegloticase mixed with saline, mice repeatedly dosed with pegloticase containing different MW or amount of free PEG possessed 4- to 12- fold lower anti-PEG IgG, and 6- to 10- fold lower anti-PEG IgM, after 3 rounds of pegloticase dosed every 2 weeks. The markedly reduced APA levels, together with competitive inhibition by free PEG, restored the prolonged circulation of pegloticase to levels observed in APA-naïve animals. In contrast, mice with pegloticase-induced APA eliminated nearly all pegloticase from the circulation within just four hours post-injection. These results support the growing literature demonstrating free PEG may effectively suppress drug-induced APA, which in turn may offer sustained therapeutic benefits without requiring broad immunomodulation. We also showed free PEG effectively blocked the PEGylated protein from binding with cells expressing PEG-specific B cell receptors. It provides a template of how we may be able to tune the interactions and immunogenicity of other polymer-modified therapeutics. STATEMENT OF SIGNIFICANCE: A major challenge with engineering materials for drug delivery is their interactions with the immune system. For instance, our body can produce high levels of anti-PEG antibodies (APA). Unfortunately, the field currently lack tools to limit immunostimulation or overcome pre-existing anti-PEG antibodies, without using broad immunosuppression. Here, we showed that simply introducing free PEG into a clinical formulation of PEG-uricase can effectively limit induction of anti-PEG antibodies, and restore their prolonged circulation upon repeated dosing. Our work offers a readily translatable method to safely and effectively restore the use PEG-drugs in patients with PEG-immunity, and provides a template to use unconjugated polymers with low immunogenicity to regulate interactions with the immune system for other polymer-modified therapeutics.


Antibodies , Urate Oxidase , Humans , Animals , Mice , Molecular Weight , Urate Oxidase/therapeutic use , Antibodies/pharmacology , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use
10.
Nat Rev Rheumatol ; 19(10): 640-649, 2023 Oct.
Article En | MEDLINE | ID: mdl-37684360

Xanthine oxidase inhibitors such as allopurinol and febuxostat have been the mainstay urate-lowering therapy (ULT) for treating hyperuricaemia in patients with gout. However, not all patients receiving oral ULT achieve the target serum urate level, in part because some patients cannot tolerate, or have actual or misconceived contraindications to, their use, mainly due to comorbidities. ULT dosage is also limited by formularies and clinical inertia. This failure to sufficiently lower serum urate levels can lead to difficult-to-treat or uncontrolled gout, usually due to poorly managed and/or under-treated gout. In species other than humans, uricase (urate oxidase) converts urate to allantoin, which is more soluble in urine than uric acid. Exogenic uricases are an exciting therapeutic option for patients with gout. They can be viewed as enzyme replacement therapy. Uricases are being used to treat uncontrolled gout, and can achieve rapid reduction of hyperuricaemia, dramatic resolution of tophi, decreased chronic joint pain and improved quality of life. Availability, cost and uricase immunogenicity have limited their use. Uricases could become a leading choice in severe and difficult-to-treat gout as induction and/or debulking therapy (that is, for lowering of the urate pool) to be followed by chronic oral ULT. This Review summarizes the evidence regarding available uricases and those in the pipeline, their debulking effect and their outcomes related to gout and beyond.


Gout , Hyperuricemia , Humans , Gout Suppressants/therapeutic use , Gout Suppressants/adverse effects , Uric Acid , Hyperuricemia/complications , Hyperuricemia/drug therapy , Urate Oxidase/therapeutic use , Quality of Life , Gout/drug therapy , Allopurinol/therapeutic use , Allopurinol/adverse effects
11.
Biochem Biophys Res Commun ; 666: 115-121, 2023 07 23.
Article En | MEDLINE | ID: mdl-37182286

Gout is a form of inflammatory arthritis that results from elevated serum uric acid levels and the deposition of urate crystals in multiple joints. The inflammatory response during an acute gout attack is mediated by the activation of the NLRP3 inflammasome, leading to the release of IL-1ß and inducing a localized tissue inflammatory response. Urate lowering therapies such as Pegloticase effectively reduce serum uric acid levels but are generally associated with an increase in acute gout flares. In this study, we developed a long-acting anti-inflammatory recombinant uricase by sequential fusing interleukin-1 receptor antagonist (IL-1Ra) and albumin-binding domain (ABD) with the N-terminal end of Arthrobacter globiformis uricase (AgUox). The recombinant uricase has longer in vivo half-life, and significantly alleviates monosodium urate (MSU) crystals induced inflammation in mouse model compared with the wild-type AgUox. This long-acting anti-inflammatory recombinant uricase has the potential to be developed as an effective urate lowering therapy with better safety profiles.


Arthritis, Gouty , Gout , Animals , Mice , Uric Acid , Half-Life , Urate Oxidase/genetics , Urate Oxidase/therapeutic use , Gout/drug therapy , Anti-Inflammatory Agents/therapeutic use , Inflammasomes
12.
J Oncol Pharm Pract ; 29(4): 893-898, 2023 Jun.
Article En | MEDLINE | ID: mdl-35306913

Purpose: Hyperuricemia is a complication arising from tumor lysis syndrome (TLS). Literature has shown that a single 3 mg dose was just as efficacious as a single 6 mg dose when the uric acid (UA) levels were ≤12 mg/dL. Here, we present a multi-center analysis rasburicase utilization and its effect on healthcare costs. Methods: This is a multi-center, retrospective analysis of adult cancer patients who were admitted to Methodist Le Bonheur Healthcare hospitals and received rasburicase from February 2020 to February 2021. The primary endpoint was to test whether rasburicase 3 mg had similar rates of uric acid normalization (defined as uric acid ≤7.5 mg/dL) within 24 h as a dose of 6 mg. Results: Seventy-nine patients were included in the study. While the baseline uric acid was lower in the 3 mg arm compared to the 6 mg arms, there was no difference in the uric acid normalization at 24 h between the 3 mg arm (95%) and 6 mg arm (82%) (p = 0.134). A cost-savings of over $300,000 annually can be achieved with the proposed protocol. Conclusion: A single, fixed rasburicase dose of 3 mg was effective in normalizing uric acid levels within 24 h, and is associated with significant cost-savings.


Hyperuricemia , Tumor Lysis Syndrome , Adult , Humans , Tumor Lysis Syndrome/etiology , Gout Suppressants/adverse effects , Uric Acid , Retrospective Studies , Urate Oxidase/therapeutic use , Hyperuricemia/drug therapy , Hyperuricemia/etiology
13.
Arthritis Res Ther ; 24(1): 281, 2022 12 27.
Article En | MEDLINE | ID: mdl-36575505

BACKGROUND: Uncontrolled/refractory gout patients are recalcitrant/intolerant to oral urate-lowering therapies (ULTs), experiencing frequent gout flares, functionally limiting tophi, and low quality of life. Pegloticase lowers urate, but anti-pegloticase antibodies limit urate-lowering efficacy and increase infusion reaction (IR) risk. Immunomodulator + pegloticase co-administration may improve treatment response rates, with 79% of MIRROR open-label trial (MIRROR-OL, pegloticase + oral methotrexate) participants meeting 6-month response criteria. Exploratory outcomes from MIRROR-OL are described here. METHODS: Adults with uncontrolled gout (serum urate [SU] ≥ 6 mg/dL and ULT-intolerance/recalcitrance or functionally limiting tophi) were included. Oral methotrexate (15 mg/week) was administered 4 weeks before and during pegloticase treatment (biweekly 8 mg infusion, ≤ 52 weeks). Exploratory outcomes included change from baseline (CFB) in number of affected joints, Health Assessment Questionnaires (HAQs), and Gout Global Assessments. RESULTS: Fourteen patients received ≥ 1 pegloticase infusion, with 13 included in 52-week analyses (1 enrolled before treatment-extension amendment, exited at 24 weeks). Three patients prematurely exited due to SU rise; 10 completed 52-week evaluations (8 completed 52 weeks of co-therapy, 2 completed 24 weeks [met treatment goals]). At 52 weeks, SU averaged 1.1 ± 2.5 mg/dL, with improvements in HAQ pain and health (CFB: - 33.6 and - 0.7, respectively), Patient and Physician Global Assessments (CFB: - 4.6 and - 5.7, respectively), and joint involvement (CFB: - 5.6, - 8.4, - 6.0 tender, swollen, tophi-affected joints, respectively). Two patients underwent dual-energy computed tomography, showing concomitant monosodium urate volume reductions. All patients had ≥ 1 AE, with 92.9% experiencing acute flare. One mild IR ("cough") occurred and no new safety signals were identified. CONCLUSION: Pegloticase + methotrexate co-therapy resulted in sustained SU-lowering with meaningful improvements in clinical measures, urate burden, and patient-reported outcomes. TRIAL REGISTRATION: ClinicalTrials.gov (NCT03635957).


Gout , Uric Acid , Adult , Humans , Methotrexate/therapeutic use , Gout Suppressants/therapeutic use , Quality of Life , Gout/drug therapy , Gout/chemically induced , Polyethylene Glycols/adverse effects , Urate Oxidase/therapeutic use , Treatment Outcome
14.
Int J Rheum Dis ; 25(12): 1395-1407, 2022 Dec.
Article En | MEDLINE | ID: mdl-36082436

AIM: Jian Pi Shen Shi Formula (JPSSF) is a beneficial treatment for hyperuricemia and related tissue damage in the clinical setting. This study was designed to investigate its therapeutic potential and underlying mechanisms in uricase-deficient rats (Uox-/- rats). METHODS: Uox-/- rats were used to assess the therapeutic potential of JPSSF on hyperuricemia. Protein extracts from renal tissues of a Uox-/- group and a JPSSF group were analyzed using tandem mass tag labeling quantitative proteomic workflow. Collagen deposition in Uox-/- rat kidneys was analyzed by Masson trichromatic staining. The gene expression associated with collagen-binding-related signaling pathways in the kidneys was further explored using quantitative polymerase chain reaction assay. The protein expressions of collagen 1a1 (col1a1), col6a1, and α-smooth muscle actin were measured by Western blotting and immunohistochemistry. RESULTS: JPSSF significantly decreased renal function indices and alleviated renal injuries. The action of JPSSF was manifested by down-regulation of col6a1 and interleukin-1 receptor-associated kinase-like 2, which blocked the binding sites on collagen and further prevented kidney injury. The anti-renal fibrosis effect of JPSSF was confirmed by reducing the collagen deposition and hydroxyproline concentrations. JPSSF treatment also intensely down-regulated the mRNA and protein expressions of col6a1, col1a1, and α-smooth muscle actin, which inhibited the function of the collagen-binding-related signaling pathway. CONCLUSION: Our results indicated that JPSSF notably ameliorated hyperuricemia and related renal fibrosis in Uox-/- rats through lowering uric acid and down-regulating the function of the collagen-binding pathway. This suggested that JPSSF is a potential empirical formula for treating hyperuricemia and accompanying renal fibrosis.


Hyperuricemia , Kidney Diseases , Rats , Animals , Hyperuricemia/complications , Hyperuricemia/drug therapy , Urate Oxidase/metabolism , Urate Oxidase/pharmacology , Urate Oxidase/therapeutic use , Actins/metabolism , Proteomics , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Kidney Diseases/prevention & control , Fibrosis , Kidney/pathology , Signal Transduction , Collagen/metabolism
15.
J Physiol Biochem ; 78(3): 679-687, 2022 Aug.
Article En | MEDLINE | ID: mdl-35674867

Conventional chemotherapy plays a key role in hepatocellular carcinoma (HCC) treatment, however, with intrinsic or acquired chemoresistance being a major constraint. Here, we aimed to identify potential target to reverse such chemoresistance. In the present study, we found significant difference in uridine monophosphate synthetase (UMPS) expression between 5-FU resistant and sensitive HCC cell lines and the overexpression or downregulation of UMPS impacted 5-FU response in HCC cells. We further found that inhibition of UMPS activity with uric acid at concentration present in human plasma decreased the 5-FU sensitivity of HCC cells, while reduction of uric acid levels with uricase improved the 5-FU sensitivity of HCC cells as well as colorectal cancer cells. In vivo studies also suggested that modulation of uric acid levels did affect 5-FU sensitivity of tumors. These data indicated that UMPS was correlated with the 5-FU resistance in HCC cells and uricase sensitized cancer cells to 5-FU through uricase-uric acid-UMP synthase axis, which provided a potential strategy to improve the efficacy of 5-FU-based chemotherapy for human cancers.


Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Liver Neoplasms/metabolism , Multienzyme Complexes , Orotate Phosphoribosyltransferase , Orotidine-5'-Phosphate Decarboxylase , Urate Oxidase/therapeutic use , Uric Acid
16.
Gut Microbes ; 14(1): 2070391, 2022.
Article En | MEDLINE | ID: mdl-35491895

Hyperuricemia is the second most prevalent metabolic disease to human health after diabetes. Only a few clinical drugs are available, and most of them have serious side effects. The human body does not have urate oxidase, and uric acid is secreted via the kidney or the intestine. Reduction through kidney secretion is often the cause of hyperuricemia. We hypothesized that the intestine secretion could be enhanced when a recombinant urate-degrading bacterium was introduced into the gut. We engineered an Escherichia coli Nissle 1917 strain with a plasmid containing a gene cassette that encoded two proteins PucL and PucM for urate metabolism from Bacillus subtilis, the urate importer YgfU and catalase KatG from E. coli, and the bacterial hemoglobin Vhb from Vitreoscilla sp. The recombinant E. coli strain effectively degraded uric acid under hypoxic conditions. A new method to induce hyperuricemia in mice was developed by intravenously injecting uric acid. The engineered Escherichia coli strain significantly lowered the serum uric acid when introduced into the gut or directly injected into the blood vessel. The results support the use of urate-degrading bacteria in the gut to treat hyperuricemia. Direct injecting bacteria into blood vessels to treat metabolic diseases is proof of concept, and it has been tried to treat solid tumors.


Escherichia coli Infections , Gastrointestinal Microbiome , Hyperuricemia , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Hyperuricemia/drug therapy , Mice , Oxygen , Urate Oxidase/genetics , Urate Oxidase/therapeutic use , Uric Acid/metabolism , Uric Acid/therapeutic use
17.
BioDrugs ; 36(2): 95-103, 2022 Mar.
Article En | MEDLINE | ID: mdl-35316517

Refractory, or uncontrolled, gout is a chronic, progressive, inflammatory arthropathy resulting from continued urate deposition after failed attempts to lower serum uric acid below the therapeutic threshold with oral urate-lowering therapies such as allopurinol and febuxostat. Recombinant uricase is increasingly being used to treat refractory gout; however, the immunogenicity of uricase-based therapies has limited the use of these biologic therapies. Antidrug antibodies against biologic therapies, including uricase and PEGylated uricase, can lead to loss of urate-lowering response, increased risk of infusion reactions, and subsequent treatment failure. However, co-therapy with an immunomodulator can attenuate antidrug antibody development, potentially increasing the likelihood of sustained urate lowering, therapy course completion, and successful treatment outcomes. This review summarizes evidence surrounding the use of immunomodulation as co-therapy with recombinant uricases.


Gout , Uric Acid , Allopurinol/therapeutic use , Antibodies/therapeutic use , Gout/drug therapy , Gout Suppressants/adverse effects , Gout Suppressants/therapeutic use , Humans , Urate Oxidase/therapeutic use , Uric Acid/therapeutic use
18.
Adv Sci (Weinh) ; 9(11): e2103672, 2022 04.
Article En | MEDLINE | ID: mdl-35133079

Protein therapeutics, except for antibodies, have a short plasma half-life and poor stability in circulation. Covalent coupling of polyethylene glycol (PEG) to protein drugs addresses this limitation. However, unlike previously thought, PEG is immunogenic. In addition to induced PEG antibodies, ≈70% of the US population has pre-existing anti-PEG antibodies. Both induced and preexisting anti-PEG antibodies result in accelerated drug clearance, reduced clinical efficacy, and severe hypersensitivity reactions that have limited the clinical utility of uricase, an enzyme drug for treatment for refractory gout that is decorated with a PEG corona. Here, the authors synthesize a poly(oligo(ethylene glycol) methyl ether methacrylate) (POEGMA) conjugate of uricase that decorates the protein with multiple polymer chains to create a corona to solve these problems. The resulting uricase-POEGMA is well-defined, has high bioactivity, and outperforms its PEG counterparts in its pharmacokinetics (PK). Furthermore, the conjugate does not induce anti-POEGMA antibodies and is not recognized by anti-PEG antibodies. These findings suggest that POEGMA conjugation may provide a solution to the immunogenicity and antigenicity limitations of PEG while improving upon its PK benefits. These results transcend uricase and can be applied to other PEGylated therapeutics and the broader class of biologics with suboptimal PK.


Gout , Urate Oxidase , Antibodies/metabolism , Antigens/therapeutic use , Gout/drug therapy , Humans , Immunity , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/therapeutic use , Polymers/therapeutic use , Urate Oxidase/pharmacokinetics , Urate Oxidase/therapeutic use
19.
Nat Commun ; 13(1): 272, 2022 01 12.
Article En | MEDLINE | ID: mdl-35022448

Biologic drugs have transformed the standard of care for many diseases. However, many biologics induce the formation of anti-drug antibodies (ADAs), which can compromise their safety and efficacy. Preclinical studies demonstrate that biodegradable nanoparticles-encapsulating rapamycin (ImmTOR), but not free rapamycin, mitigate the immunogenicity of co-administered biologic drugs. Here we report the outcomes from two clinical trials for ImmTOR. In the first ascending dose, open-label study (NCT02464605), pegadricase, an immunogenic, pegylated uricase enzyme derived from Candida utilis, is assessed for safety and tolerability (primary endpoint) as well as activity and immunogenicity (secondary endpoint); in the second single ascending dose Phase 1b trial (NCT02648269) composed of both a double-blind and open-label parts, we evaluate the safety of ImmTOR (primary endpoint) and its ability to prevent the formation of anti-drug antibodies against pegadricase and enhance its pharmacodynamic activity (secondary endpoint) in patients with hyperuricemia. The combination of ImmTOR and pegadricase is well tolerated. ImmTOR inhibits the development of uricase-specific ADAs in a dose-dependent manner, thus enabling sustained enzyme activity and reduction in serum uric acid levels. ImmTOR may thus represent a feasible approach for preventing the formation of ADAs to a broad range of immunogenic biologic therapies.


Antibodies/therapeutic use , Hyperuricemia/drug therapy , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Polyethylene Glycols/pharmacology , Urate Oxidase/therapeutic use , Adult , Aged , Antibodies/chemistry , Biological Therapy , Double-Blind Method , Female , Humans , Male , Middle Aged , Organic Chemicals/chemistry , Urate Oxidase/pharmacology , Uric Acid , Young Adult
20.
Semin Nephrol ; 42(6): 151342, 2022 11.
Article En | MEDLINE | ID: mdl-37167817

Tumor lysis syndrome (TLS) and high-dose methotrexate (HD MTX) toxicity can present with potentially severe complications, including acute kidney injury, in patients with malignancy. Guidelines for using rasburicase and glucarpidase as rescue therapies for TLS and HD MTX toxicity, respectively, are widely used by clinicians intending to mitigate organ toxicity and decrease morbidity and mortality as a consequence of cancer therapy. This review discusses the pathogenesis of TLS and HD MTX-associated toxicity, to understand the mechanism of action of these therapeutic agents and to review the currently available evidence supporting their use.


Acute Kidney Injury , Tumor Lysis Syndrome , Humans , gamma-Glutamyl Hydrolase/therapeutic use , Urate Oxidase/therapeutic use , Tumor Lysis Syndrome/drug therapy , Tumor Lysis Syndrome/complications , Acute Kidney Injury/drug therapy , Acute Kidney Injury/complications
...