Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Toxicol Appl Pharmacol ; 353: 67-73, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29908244

ABSTRACT

Uridine triacetate has been shown to be an effective antidote against mortality and toxicity caused by either overdoses or exaggerated susceptibility to the widely used anticancer agents 5-fluorouracil (5-FU) and capecitabine. However, a direct assessment of efficacy based on when emergency treatment was initiated was not clinically feasible. In this study we used mouse models of 5-FU overdose and of dihydropyrimidine dehydrogenase (DPD) deficiency to compare the efficacy of uridine triacetate in reducing toxicity and mortality when treatment was initiated at time points from 4 to 144 h after administration of 5-FU. We found that uridine triacetate was effective both in the 5-FU overdose and DPD deficiency models. Starting treatment within 24 h was most effective at reducing toxicity and mortality in both models, while treatment starting more than 96 to 120 h after 5-FU was far less effective. Uridine triacetate also reduced mortality in the DPD deficiency model when mice were treated with the 5-FU prodrug capecitabine. The results of this study are supportive of clinical observations and practice, indicating that efficacy declined progressively with later and later treatment initiation. Prompt treatment with uridine triacetate, within 24 h, conferred the greatest protection against 5-FU overexposure.


Subject(s)
Acetates/therapeutic use , Antimetabolites, Antineoplastic/toxicity , Capecitabine/toxicity , Dihydropyrimidine Dehydrogenase Deficiency/drug therapy , Fluorouracil/toxicity , Uridine/analogs & derivatives , Animals , Antidotes , Antimetabolites, Antineoplastic/pharmacokinetics , Dihydropyrimidine Dehydrogenase Deficiency/chemically induced , Dihydropyrimidine Dehydrogenase Deficiency/metabolism , Dose-Response Relationship, Drug , Drug Overdose/drug therapy , Female , Fluorouracil/pharmacokinetics , Mice , Survival Analysis , Uridine/therapeutic use
2.
Cancer ; 123(2): 345-356, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27622829

ABSTRACT

BACKGROUND: Increased susceptibility to 5-fluorouracil (5-FU)/capecitabine can lead to rapidly occurring toxicity caused by impaired clearance, dihydropyrimidine dehydrogenase deficiency, and other genetic variations in the enzymes that metabolize 5-FU. Life-threatening 5-FU overdoses occur because of infusion pump errors, dosage miscalculations, and accidental or suicidal ingestion of capecitabine. Uridine triacetate (Vistogard) was approved in 2015 for adult and pediatric patients who exhibit early-onset severe or life-threatening 5-FU/capecitabine toxicities or present with an overdose. Uridine triacetate delivers high concentrations of uridine, which competes with toxic 5-FU metabolites. METHODS: In 2 open-label clinical studies, patients who presented with a 5-FU/capecitabine overdose or an early onset of severe toxicities were treated. Patients received uridine triacetate as soon as possible (most within the first 96 hours after 5-FU/capecitabine). Outcomes included survival, resumption of chemotherapy, and safety. Their survival was compared with the survival of a historical cohort of overdose patients who received only supportive care. RESULTS: A total of 137 of 142 overdose patients (96%) treated with uridine triacetate survived and had a rapid reversal of severe acute cardiotoxicity and neurotoxicity; in addition, mucositis and leukopenia were prevented, or the patients recovered from them. In the historical cohort, 21 of 25 patients (84%) died. Among the 141 uridine triacetate-treated overdose patients with a diagnosis of cancer (the noncancer patients included 6 intentional or accidental pediatric overdoses), 53 resumed chemotherapy in < 30 days (median time after 5-FU, 19.6 days), and this indicated a rapid recovery from toxicity. Adverse reactions in patients receiving uridine triacetate included vomiting (8.1%), nausea (4.6%), and diarrhea (3.5%). CONCLUSIONS: In these studies, uridine triacetate was a safe and effective lifesaving antidote for capecitabine and 5-FU overexposure, and it facilitated the rapid resumption of chemotherapy. Cancer 2017;123:345-356. © 2016 American Cancer Society.


Subject(s)
Acetates/therapeutic use , Antimetabolites, Antineoplastic/adverse effects , Capecitabine/adverse effects , Drug Overdose/drug therapy , Fluorouracil/adverse effects , Uridine/analogs & derivatives , Capecitabine/therapeutic use , Female , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Neoplasms/drug therapy , Uridine/therapeutic use
3.
J Alzheimers Dis ; 36(4): 637-57, 2013.
Article in English | MEDLINE | ID: mdl-23648515

ABSTRACT

Uridine prodrug PN401 has been shown to have neuroprotective effects in models of Parkinson's disease and Huntington's disease. These age-related neurodegenerative diseases including Alzheimer's disease (AD) are associated with mitochondrial dysfunction, oxidative stress, and inflammation. Attenuation of these pathological factors in AD, in addition to amyloid fibrils and neurofibrillary tangles, is critical to prevent cognitive impairment. The effects of PN401 treatment were tested in the Tg2576 and Tg2576 X P301L (TAPP) mouse models of AD. Treatment with PN401 reduced impairments in the Tg2576 mice in contextual fear conditioning and novel object recognition. In the TAPP mice, PN401 reduced the impairments in novel object recognition and social transmission of food preference. PN401 also improved motor behavior and reduced anxiety-like behavior in the TAPP mice. TAPP mouse hippocampal tau phosphorylation and lipid peroxidation were reduced by PN401 treatment. Increased tau phosphorylation was significantly correlated with worsening novel object recognition memory. PN401 did not affect amyloid plaque area in the AD mice. In other AD-related animal studies, PN401 treatment reduced blood-brain barrier damage due to intracortical LPS, elevation of serum TNFα due to systemic LPS, and hippocampal CA1 neuronal loss in the gerbil stroke model. Uridine dose-dependently protected cells from chemical hypoxia and ceramide, and decreased formation of reactive oxygen species and mitochondrial DNA damage due to hydrogen peroxide. These protective effects were achieved by raising uridine levels to at least 25-50 µM and serum uridine levels in this range in humans were obtained with oral PN401.


Subject(s)
Alzheimer Disease/drug therapy , Disease Models, Animal , Memory/drug effects , Prodrugs/administration & dosage , Uridine/analogs & derivatives , Uridine/administration & dosage , Acetates , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cell Line, Tumor , Cricetinae , Female , Gerbillinae , Humans , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Stroke/drug therapy , Stroke/pathology
4.
J Transl Med ; 9: 203, 2011 Nov 28.
Article in English | MEDLINE | ID: mdl-22123298

ABSTRACT

BACKGROUND: The detection of insulin autoantibodies (IAA) aids in the prediction of autoimmune diabetes development. However, the long-standing, gold standard 125I-insulin radiobinding assay (RBA) has low reproducibility between laboratories, long sample processing times and requires the use of newly synthesized radiolabeled insulin for each set of assays. Therefore, a rapid, non-radioactive, and reproducible assay is highly desirable. METHODS: We have developed electrochemiluminescence (ECL)-based assays that fulfill these criteria in the measurement of IAA and anti-insulin antibodies (IA) in non-obese diabetic (NOD) mice and in type 1 diabetic individuals, respectively. Using the murine IAA ECL assay, we examined the correlation between IAA, histopathological insulitis, and blood glucose in a cohort of female NOD mice from 4 up to 36 weeks of age. We developed a human IA ECL assay that we compared to conventional RBA and validated using samples from 34 diabetic and 59 non-diabetic individuals in three independent laboratories. RESULTS: Our ECL assays were rapid and sensitive with a broad dynamic range and low background. In the NOD mouse model, IAA levels measured by ECL were positively correlated with insulitis severity, and the values measured at 8-10 weeks of age were predictive of diabetes onset. Using human serum and plasma samples, our IA ECL assay yielded reproducible and accurate results with an average sensitivity of 84% at 95% specificity with no statistically significant difference between laboratories. CONCLUSIONS: These novel, non-radioactive ECL-based assays should facilitate reliable and fast detection of antibodies to insulin and its precursors sera and plasma in a standardized manner between laboratories in both research and clinical settings. Our next step is to evaluate the human IA assay in the detection of IAA in prediabetic subjects or those at risk of type 1 diabetes and to develop similar assays for other autoantibodies that together are predictive for the diagnosis of this common disorder, in order to improve prediction and facilitate future therapeutic trials.


Subject(s)
Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/diagnosis , Electrochemistry/methods , Insulin Antibodies/blood , Insulin-Secreting Cells/pathology , Luminescent Measurements/methods , Animals , Autoantibodies/blood , Diabetes Mellitus, Type 1/pathology , Disease Progression , Female , Humans , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred NOD , ROC Curve , Radioligand Assay , Reproducibility of Results , Sensitivity and Specificity
5.
Neurobiol Dis ; 24(3): 455-65, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17011205

ABSTRACT

Previously, uridine pro-drug 2',3',5'-tri-O-acetyluridine (PN401) was shown to be protective in the mitochondrial complex II inhibitor 3-nitropropionic acid model of Huntington's disease (HD). In this study, PN401 increased survival and improved motor function on the rotarod in both R6/2 and N171-82Q polyglutamine repeat mouse models of HD. PN401 significantly decreased neurodegeneration in both the piriform cortex and striatum although PN401 decreased huntingtin protein aggregates only in the striatum. Cortical and striatal brain-derived neurotrophic factor (BDNF) protein levels were reduced in the +/- compared to the -/- N171-82Q mice and PN401 treatment significantly increased cortical BDNF in both +/- and -/- mice, but PN401 did not affect striatal BDNF. These results suggest that PN401 may have beneficial effects in the treatment of neurodegenerative diseases such as HD.


Subject(s)
Huntington Disease/prevention & control , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Prodrugs/pharmacology , Uridine/analogs & derivatives , Acetates , Administration, Oral , Analysis of Variance , Animals , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Female , Huntingtin Protein , Huntington Disease/drug therapy , Huntington Disease/mortality , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Neurologic Mutants , Mice, Transgenic , Motor Activity/drug effects , Neostriatum/cytology , Neostriatum/drug effects , Neostriatum/metabolism , Nerve Degeneration/drug therapy , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Nuclear Proteins/drug effects , Nuclear Proteins/metabolism , Rotarod Performance Test , Uridine/administration & dosage , Uridine/pharmacology
6.
Cancer Chemother Pharmacol ; 58(1): 136-42, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16187114

ABSTRACT

PURPOSE: PN401, an oral prodrug of uridine yields more bioavailable uridine than oral administration of uridine itself. PN401 may therefore be useful for permitting dose escalation of 5-fluorouracil (5-FU) with consequent improvements in antitumor efficacy. EXPERIMENTAL DESIGN: Female BALB/c mice (Colon 26 adenocarcinoma) were treated with 5-FU with PN401 to define the MTD, and pharmacokinetic analyses were done. A comparison of 5-FU/PN401 was made to 5-FU/eniluracil (EU) and 5-FU/LV. The best timing of the first dose of PN401 relative to 5-FU was evaluated by administering groups of mice PN401 beginning 2, 24, or 48 h after 5-FU dose. RESULTS: The MTD of 5-FU was 100 mg/kg/week whereas the MTD of 5-FU + PN401 was 200 mg/kg/week. A complete response (CR) of 80% and partial response (PR) of 20% was observed with 5-FU (200 mg/kg) + PN401, CR of 40% and PR of 60% with 5-FU (175 mg/kg) + PN401, PR of 10% with 5-FU (150 mg/kg) + PN401 while no response with 5-FU (100 mg/kg) + PN401. Analysis of 5-FU pharmacokinetics displayed nonlinearity as a function of administered dose in mice. In the comparison study, the best response was achieved with PN401 when compared to EU and LV. Mice that did not receive PN401 died by day 12, while other groups were alive at day 31. The proportion of mice surviving was highest in the group which received PN401 at 2 h followed by 24 and 48 h. CONCLUSIONS: There is a threshold 5-FU dose after which the efficacy is dramatically improved-in mice bearing Colon 26 adenocarcinoma, that threshold is a dose of >150 mg/kg/week, and the increased efficacy correlates with about a fourfold increase in the AUC of 5-FU. PN401 used to rescue mice from the lethal toxicity of 5-FU entails that PN401 can be used as an antidote even when used up to 48 h after a 5-FU overdose.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , Acetates , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Humans , Leucovorin/administration & dosage , Maximum Tolerated Dose , Mice , Mice, Inbred BALB C , Survival Rate , Tumor Burden/drug effects , Uridine/administration & dosage , Uridine/analogs & derivatives , Xenograft Model Antitumor Assays
7.
Brain Res ; 1066(1-2): 164-71, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16330000

ABSTRACT

It has been hypothesized that mitochondrial respiratory chain dysfunction leads to a pyrimidine deficiency since the pyrimidine biosynthetic enzyme dihydroorotate dehydrogenase is coupled to the electron transport chain. The uridine prodrug triacetyluridine (PN401) is neuroprotective in several models of neurodegenerative disease involving respiratory chain toxins. Therefore, the therapeutic effects of PN401 might involve the correction of a pyrimidine deficiency secondary to respiratory chain impairment. We infused mice with the cytochrome c oxidase inhibitor azide, which inhibited brain complex IV activity. Chronic infusion of azide for 2 or 14 days induced significant toxicity and mortality but did not cause a pyrimidine deficit in the brain. In contrast, the pyrimidine synthesis inhibitor N-phosphonoacetyl-l-aspartate (PALA) produced a pyrimidine deficit with minimal mortality. Treatment with 6% PN401 decreased mortality and cerebrocortical apoptosis caused by azide. Previously, we found that optimal neuroprotection against mitochondrial complex II inhibition required 4-6% PN401. PN401 at 1, 3, 6 and 10% in chow induced nonlinear increases in plasma uridine with 6% PN401 elevating plasma uridine up to 80 muM, and these higher micromolar uridine levels were also required for neuroprotection in chemical hypoxia models in vitro. Our results indicate that severe complex IV inhibition in vivo does not lead to a pyrimidine deficiency, and therefore the protective effect of PN401 in the azide toxin model is not mediated through the correction of a pyrimidine deficiency. Furthermore, supraphysiological levels of uridine are required to produce optimal protective effects in disorders involving impairment of mitochondrial respiratory complex II or IV.


Subject(s)
Electron Transport Complex IV/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neuroprotective Agents , Prodrugs/pharmacology , Pyrimidines/metabolism , Uridine/analogs & derivatives , Uridine/pharmacology , Acetates , Animals , Apoptosis/drug effects , Aspartic Acid/analogs & derivatives , Aspartic Acid/pharmacology , Azides/antagonists & inhibitors , Azides/toxicity , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Electron Transport/drug effects , Fibroblasts/metabolism , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Phosphonoacetic Acid/analogs & derivatives , Phosphonoacetic Acid/pharmacology , Prodrugs/administration & dosage , Uridine/administration & dosage , Uridine/metabolism
8.
Neuromolecular Med ; 6(2-3): 87-92, 2004.
Article in English | MEDLINE | ID: mdl-15970626

ABSTRACT

Administration of triacetyluridine (TAU) is a means of delivering exogenous pyrimidines to the brain, which may help to compensate for bioenergetic defects. TAU has previously been shown to be neuroprotective in animal models of Huntington's and Alzheimer's diseases. We examined whether oral administration of TAU in the diet could exert significant neuroprotective effects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxicity model of Parkinson's disease. Administration of TAU significantly attenuated MPTP-induced depletion of striatal dopamine and loss of tyrosine-hydroxylase-positive neurons in the substantia nigra. These findings suggest that administration of TAU may be a novel approach for treating neurodegenerative diseases associated with impaired mitochondrial function.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacokinetics , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Uridine/analogs & derivatives , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 3,4-Dihydroxyphenylacetic Acid/metabolism , Acetates , Animals , Dietary Supplements , Disease Models, Animal , Dopamine/metabolism , Homovanillic Acid/metabolism , MPTP Poisoning/prevention & control , Mice , Neurotoxins/antagonists & inhibitors , Uridine/administration & dosage , Uridine/pharmacology
9.
Brain Res ; 994(1): 44-54, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14642447

ABSTRACT

Huntington's disease (HD) is associated with decreased activity of mitochondrial succinate dehydrogenase (complex II). De novo biosynthesis of uridine nucleotides is directly coupled to the respiratory chain. Cells with impaired mitochondrial function become uridine auxotrophs and can be maintained with high micromolar concentration of uridine and pyruvate. The therapeutic role of pyrimidines and possible changes in uridine content has not been assessed in neurological diseases involving mitochondrial dysfunction in vivo. Oral administration of PN401 delivers much higher levels of uridine to the circulation than oral administration of uridine itself. Administration of complex II inhibitor 3-nitropropionic acid (3NP) induced neuronal damage in the striatum, substantia nigra and/or thalamus in 80% of the mice and led to 38% mortality. Treatment with PN401 almost completely prevented the neuronal damage due to 3NP and completely prevented mortality. In two subsequent experiments, 3NP-induced weight loss, mortality and behavioral impairment in rotarod performance and spontaneous motor activity were attenuated by treatment with oral PN401. 3NP did not reduce forebrain total uridine nucleotides (TUN), though higher doses of PN401 associated with optimal neuroprotection did elevate TUN to supranormal levels. Thus, oral PN401 treatment has neuroprotective effects in a HD model of mitochondrial dysfunction and the mechanism is more complex than correction of a pyrimidine deficit.


Subject(s)
Huntington Disease/drug therapy , Nerve Degeneration/drug therapy , Prodrugs/therapeutic use , Propionates/toxicity , Uridine/analogs & derivatives , Uridine/therapeutic use , Acetates , Administration, Oral , Animals , Huntington Disease/chemically induced , Huntington Disease/pathology , Male , Mice , Mitochondria/drug effects , Mitochondria/pathology , Motor Activity/drug effects , Motor Activity/physiology , Nerve Degeneration/pathology , Nitro Compounds , Prodrugs/pharmacology , Uridine/pharmacology , Weight Loss/drug effects , Weight Loss/physiology
SELECTION OF CITATIONS
SEARCH DETAIL