Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 361
Filtrar
1.
Nat Rev Dis Primers ; 10(1): 58, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39174582

RESUMEN

Tumour lysis syndrome (TLS) represents a critical oncological emergency characterized by extensive tumour cell breakdown, leading to the swift release of intracellular contents into the systemic circulation, outpacing homeostatic mechanisms. This process results in hyperuricaemia (a by-product of intracellular DNA release), hyperkalaemia, hyperphosphataemia, hypocalcaemia and the accumulation of xanthine. These electrolyte and metabolic imbalances pose a significant risk of acute kidney injury, cardiac arrhythmias, seizures, multiorgan failure and, rarely, death. While TLS can occur spontaneously, it usually arises shortly after the initiation of effective treatment, particularly in patients with a large cancer cell mass (defined as ≥500 g or ≥300 g/m2 of body surface area in children). To prevent TLS, close monitoring and hydration to improve renal perfusion and urine output and to minimize uric acid or calcium phosphate precipitation in renal tubules are essential. Intervention is based on the risk of a patient of having TLS and can include rasburicase and allopurinol. Xanthine, typically enzymatically converted to uric acid, can accumulate when xanthine oxidases, such as allopurinol, are administered during TLS management. Whether measurement of xanthine is clinically useful to optimize the use of allopurinol or rasburicase remains to be determined.


Asunto(s)
Alopurinol , Síndrome de Lisis Tumoral , Síndrome de Lisis Tumoral/fisiopatología , Síndrome de Lisis Tumoral/etiología , Síndrome de Lisis Tumoral/diagnóstico , Síndrome de Lisis Tumoral/complicaciones , Humanos , Alopurinol/uso terapéutico , Hiperuricemia/fisiopatología , Hiperuricemia/complicaciones , Urato Oxidasa/uso terapéutico , Hiperpotasemia/fisiopatología , Hiperpotasemia/etiología , Hiperpotasemia/terapia , Ácido Úrico , Xantina , Neoplasias/fisiopatología , Neoplasias/complicaciones
2.
J Control Release ; 372: 862-873, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38906421

RESUMEN

Improving the activity of uricase and lowering its immunogenicity remain significant challenges in the enzyme replacement management of hyperuricemia and related inflammatory diseases. Herein, an immunogenicity-masking strategy based on engineered red blood cells (RBCs) was developed for effective uricase delivery against both hyperuricemia and gout. The dynamic membrane of RBCs enabled high resistance to protease inactivation and hydrogen peroxide accumulation. Benefiting from these advantages, a single infusion of RBC-loaded uricase (Uri@RBC) performed prolonged blood circulation and sustained hyperuricemia management. Importantly, RBCs masked the immunogenicity of uricase, leading to the maintenance of UA-lowering performance after repeated infusion through reduced antibody-mediated macrophage clearance. In an acute gout model, Uri@RBC profoundly alleviated joint edema and inflammation with minimal systemic toxicity. This study supports the employment of immunogenicity-masking tools for efficient and safe enzyme delivery, and this strategy may be leveraged to improve the usefulness of enzyme replacement therapies for managing a wide range of inflammatory diseases.


Asunto(s)
Eritrocitos , Gota , Hiperuricemia , Urato Oxidasa , Urato Oxidasa/administración & dosificación , Urato Oxidasa/uso terapéutico , Urato Oxidasa/inmunología , Hiperuricemia/tratamiento farmacológico , Hiperuricemia/inmunología , Animales , Gota/inmunología , Eritrocitos/inmunología , Masculino , Humanos , Ácido Úrico/sangre , Ratones , Ratones Endogámicos C57BL
3.
Arthritis Care Res (Hoboken) ; 76(10): 1361-1370, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38719773

RESUMEN

OBJECTIVE: The objective of this study was to ascertain pegloticase persistence and adverse events associated with concomitant immunomodulatory drug treatment in patients with gout. METHODS: We conducted a retrospective analysis of patients with gout using the American College of Rheumatology's Rheumatology Informatics System for Effectiveness registry from January 2016 through June 2020. The first pegloticase infusion defined the index date. Based on concomitant immunomodulatory drug treatment, we identified three exposure groups: (1) immunomodulatory drug initiators (patients initiating an immunomodulatory prescription ±60 days from the index date), (2) prevalent immunomodulatory drug recipients (patients receiving their first immunomodulatory drug prescription >60 days before the index date with at least one prescription within ±60 days of the index date), and (3) immunomodulatory nonrecipients (patients receiving pegloticase without concomitant immunomodulatory drugs). We calculated the proportion of patients who achieved serum urate levels ≤6 mg/dL and who had laboratory abnormalities (white blood cell count <3.4 x 109/L, platelet count <135,000, hematocrit level <30%, alanine aminotransferase or aspartate aminotransferase level ≥1.5 times the upper limit normal value) within 180 days after the index date. Cox regression analyzed time to pegloticase discontinuation, controlling for potential confounders. RESULTS: We identified 700 pegloticase recipients (91 immunomodulatory drug initiators, 33 prevalent immunomodulatory drug recipients, and 576 nonrecipients), with a median follow-up of 14 months. Immunomodulatory drug recipients were less likely to discontinue pegloticase. The adjusted hazard ratios of pegloticase discontinuation associated with concomitant immunomodulatory drug initiation and prevalent treatment were 0.52 (95% confidence interval [CI] 0.37-0.75) and 0.69 (95% CI 0.42-1.16), respectively. Laboratory abnormalities were uncommon (<5%) and were not higher in concomitant immunomodulatory drug treatment. CONCLUSION: Consistent with clinical trials, results from this large observational registry suggest that concomitant immunomodulatory drug treatment improves pegloticase persistence.


Asunto(s)
Gota , Sistema de Registros , Urato Oxidasa , Humanos , Estudios Retrospectivos , Masculino , Femenino , Persona de Mediana Edad , Urato Oxidasa/uso terapéutico , Anciano , Gota/tratamiento farmacológico , Gota/sangre , Resultado del Tratamiento , Agentes Inmunomoduladores/uso terapéutico , Supresores de la Gota/uso terapéutico , Supresores de la Gota/efectos adversos , Quimioterapia Combinada , Ácido Úrico/sangre , Factores Inmunológicos/uso terapéutico , Factores Inmunológicos/efectos adversos , Factores de Tiempo , Polietilenglicoles
4.
Nano Lett ; 24(22): 6634-6643, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38742828

RESUMEN

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.


Asunto(s)
Hiperuricemia , Compuestos de Manganeso , Óxidos , Urato Oxidasa , Hiperuricemia/tratamiento farmacológico , Urato Oxidasa/química , Urato Oxidasa/uso terapéutico , Urato Oxidasa/metabolismo , Óxidos/química , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Humanos , Paladio/química , Paladio/farmacología , Animales , Catálisis , Ácido Úrico/química , Ratones
5.
Int J Hematol ; 119(6): 660-666, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38575822

RESUMEN

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.


Asunto(s)
Alopurinol , Leucemia Mieloide Aguda , Síndrome de Lisis Tumoral , Urato Oxidasa , Humanos , Síndrome de Lisis Tumoral/etiología , Síndrome de Lisis Tumoral/prevención & control , Urato Oxidasa/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/complicaciones , Masculino , Femenino , Persona de Mediana Edad , Alopurinol/uso terapéutico , Alopurinol/administración & dosificación , Anciano , Adulto , Quimioterapia de Inducción , Anciano de 80 o más Años , Hiperuricemia/tratamiento farmacológico , Adolescente , Incidencia , Adulto Joven
6.
Joint Bone Spine ; 91(4): 105715, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38447697

RESUMEN

OBJECTIVE: Monosodium-urate (MSU) crystal deposits can be visualized and quantified with dual-energy CT (DECT). Pegloticase lowers serum urate (SU) in uncontrolled gout patients, with methotrexate (MTX) co-therapy recommended to increase SU-lowering response rate and decrease infusion reaction risk. The literature on serial DECT-imaging during pegloticase+MTX co-therapy is sparse, with only 2 prior cases of rapid MSU deposition depletion with subsequent bone-erosion remodeling reported from a small open-label trial. Here, we report DECT findings during pegloticase treatment in a larger number of patients from a randomized controlled trial to confirm bone-erosion remodeling that follows MSU depletion with pegloticase. The influence of length-of-therapy is also explored. METHODS: Patients received pegloticase (8mg every 2weeks)+MTX (15mg/week orally) or pegloticase+placebo (PBO) during the MIRROR RCT trial. A subset underwent DECT-imaging on Day1 (first pegloticase infusion) and at Weeks 14, 24, and 52. Patients with paired baseline-Week 52 images were included. Imaged regions with baseline MSU-crystal volume (VMSU)<0.5cm3 were excluded to minimize artifact contributions. VMSU and bone-erosion remodeling were assessed. RESULTS: Eight patients (6 MTX, 2 PBO) were included. Included patients had received 52weeks (5 MTX), 42weeks (1 PBO), and 6weeks (1 MTX, 1 PBO) of pegloticase therapy. Patients who prematurely discontinued pegloticase maintained SU<6mg/dL on allopurinol (n=2)/febuxostat (n=1). At Week 52, VMSU had markedly decreased in both the pegloticase+MTX and pegloticase+PBO treatment groups, with faster depletion during pegloticase therapy. Bone-erosion remodeling was observed in 29/42 (69%) evaluated erosions: 29 (69%) size decrease, 4 (9.5%) recortication, 3 (7.1%) new bone formation. CONCLUSION: Rapid VMSU depletion during pegloticase therapy was observed with concomitant bone remodeling within 1year. Following pegloticase discontinuation, VMSU reduction slowed or stopped even when SU was maintained<6mg/dL with oral ULT. CLINICAL TRIAL REGISTRATION: NCT03994731.


Asunto(s)
Remodelación Ósea , Supresores de la Gota , Gota , Metotrexato , Tomografía Computarizada por Rayos X , Urato Oxidasa , Ácido Úrico , Humanos , Urato Oxidasa/uso terapéutico , Ácido Úrico/sangre , Gota/tratamiento farmacológico , Gota/diagnóstico por imagen , Gota/sangre , Masculino , Supresores de la Gota/uso terapéutico , Tomografía Computarizada por Rayos X/métodos , Metotrexato/uso terapéutico , Femenino , Persona de Mediana Edad , Remodelación Ósea/efectos de los fármacos , Anciano , Quimioterapia Combinada , Resultado del Tratamiento , Polietilenglicoles
7.
Int J Mol Sci ; 25(6)2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38542302

RESUMEN

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.


Asunto(s)
Síndrome Cardiorrenal , Hiperuricemia , Síndrome de Lisis Tumoral , Humanos , Hiperuricemia/tratamiento farmacológico , Síndrome Cardiorrenal/tratamiento farmacológico , Estudios Retrospectivos , Síndrome de Lisis Tumoral/tratamiento farmacológico , Síndrome de Lisis Tumoral/etiología , Síndrome de Lisis Tumoral/prevención & control , Urato Oxidasa/uso terapéutico
8.
Nano Lett ; 24(11): 3432-3440, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38391135

RESUMEN

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.


Asunto(s)
Hiperuricemia , Peroxidasa , Humanos , Peroxidasa/uso terapéutico , Urato Oxidasa/uso terapéutico , Povidona/uso terapéutico , Hiperuricemia/tratamiento farmacológico , Peróxido de Hidrógeno , Ácido Úrico/metabolismo , Oxidorreductasas , Colorantes
9.
Rheumatology (Oxford) ; 63(4): 1058-1067, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-37449908

RESUMEN

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.


Asunto(s)
Gota , Estomatitis , Adulto , Humanos , Urato Oxidasa/uso terapéutico , Urato Oxidasa/efectos adversos , Supresores de la Gota/efectos adversos , Ácido Úrico , Resultado del Tratamiento , Brote de los Síntomas , Polietilenglicoles/efectos adversos , Uricosúricos/uso terapéutico , Estomatitis/inducido químicamente , Estomatitis/tratamiento farmacológico
10.
Drug Deliv Transl Res ; 14(7): 1820-1838, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38127247

RESUMEN

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.


Asunto(s)
Artritis Gotosa , Indometacina , Ratas Sprague-Dawley , Urato Oxidasa , Ácido Úrico , Indometacina/administración & dosificación , Animales , Urato Oxidasa/administración & dosificación , Urato Oxidasa/farmacocinética , Urato Oxidasa/uso terapéutico , Ácido Úrico/sangre , Masculino , Artritis Gotosa/tratamiento farmacológico , Nanopartículas/administración & dosificación , Ratas , Ratones , Inflamación/tratamiento farmacológico , Distribución Tisular , Lesión Renal Aguda/tratamiento farmacológico , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacocinética , Antiinflamatorios/farmacología , Sistemas de Liberación de Medicamentos , Riñón/efectos de los fármacos , Riñón/metabolismo , Humanos , Células RAW 264.7 , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacocinética , Antiinflamatorios no Esteroideos/uso terapéutico , Antiinflamatorios no Esteroideos/farmacología
11.
Curr Rheumatol Rep ; 26(3): 69-80, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38133712

RESUMEN

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.


Asunto(s)
Gota , Hiperuricemia , Humanos , Supresores de la Gota/uso terapéutico , Urato Oxidasa/uso terapéutico , Ácido Úrico , Gota/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Hiperuricemia/tratamiento farmacológico
12.
Arthritis Res Ther ; 25(1): 247, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38111075

RESUMEN

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.


Asunto(s)
Gota , Urato Oxidasa , Ratones , Animales , Ratas , Humanos , Urato Oxidasa/uso terapéutico , Leucocitos Mononucleares/metabolismo , Ratas Sprague-Dawley , Gota/tratamiento farmacológico , Supresores de la Gota/uso terapéutico , Ratones Transgénicos , Polietilenglicoles/uso terapéutico , Albúminas/uso terapéutico
13.
Nat Rev Rheumatol ; 19(10): 640-649, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37684360

RESUMEN

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.


Asunto(s)
Gota , Hiperuricemia , Humanos , Supresores de la Gota/uso terapéutico , Supresores de la Gota/efectos adversos , Ácido Úrico , Hiperuricemia/complicaciones , Hiperuricemia/tratamiento farmacológico , Urato Oxidasa/uso terapéutico , Calidad de Vida , Gota/tratamiento farmacológico , Alopurinol/uso terapéutico , Alopurinol/efectos adversos
14.
Acta Biomater ; 170: 250-259, 2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37659730

RESUMEN

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.


Asunto(s)
Anticuerpos , Urato Oxidasa , Humanos , Animales , Ratones , Peso Molecular , Urato Oxidasa/uso terapéutico , Anticuerpos/farmacología , Polietilenglicoles/farmacología , Polietilenglicoles/uso terapéutico
15.
Biochem Biophys Res Commun ; 666: 115-121, 2023 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-37182286

RESUMEN

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.


Asunto(s)
Artritis Gotosa , Gota , Animales , Ratones , Ácido Úrico , Semivida , Urato Oxidasa/genética , Urato Oxidasa/uso terapéutico , Gota/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Inflamasomas
16.
J Oncol Pharm Pract ; 29(4): 893-898, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35306913

RESUMEN

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.


Asunto(s)
Hiperuricemia , Síndrome de Lisis Tumoral , Adulto , Humanos , Síndrome de Lisis Tumoral/etiología , Supresores de la Gota/efectos adversos , Ácido Úrico , Estudios Retrospectivos , Urato Oxidasa/uso terapéutico , Hiperuricemia/tratamiento farmacológico , Hiperuricemia/etiología
17.
Arthritis Res Ther ; 24(1): 281, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36575505

RESUMEN

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).


Asunto(s)
Gota , Ácido Úrico , Adulto , Humanos , Metotrexato/uso terapéutico , Supresores de la Gota/uso terapéutico , Calidad de Vida , Gota/tratamiento farmacológico , Gota/inducido químicamente , Polietilenglicoles/efectos adversos , Urato Oxidasa/uso terapéutico , Resultado del Tratamiento
18.
Int J Rheum Dis ; 25(12): 1395-1407, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36082436

RESUMEN

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.


Asunto(s)
Hiperuricemia , Enfermedades Renales , Ratas , Animales , Hiperuricemia/complicaciones , Hiperuricemia/tratamiento farmacológico , Urato Oxidasa/metabolismo , Urato Oxidasa/farmacología , Urato Oxidasa/uso terapéutico , Actinas/metabolismo , Proteómica , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/etiología , Enfermedades Renales/prevención & control , Fibrosis , Riñón/patología , Transducción de Señal , Colágeno/metabolismo
19.
J Physiol Biochem ; 78(3): 679-687, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35674867

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Humanos , Neoplasias Hepáticas/metabolismo , Complejos Multienzimáticos , Orotato Fosforribosiltransferasa , Orotidina-5'-Fosfato Descarboxilasa , Urato Oxidasa/uso terapéutico , Ácido Úrico
20.
Gut Microbes ; 14(1): 2070391, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35491895

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli , Microbioma Gastrointestinal , Hiperuricemia , Animales , Escherichia coli/genética , Escherichia coli/metabolismo , Hiperuricemia/tratamiento farmacológico , Ratones , Oxígeno , Urato Oxidasa/genética , Urato Oxidasa/uso terapéutico , Ácido Úrico/metabolismo , Ácido Úrico/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA