210589-09-6
Mucopolysaccharidosis Type I (MPS I)
[Uridine triacetate is a pyrimidine analog with a unique dual-indication profile, functioning as both an orphan drug for a rare genetic metabolic disorder and a critical emergency antidote in oncology. Identified by DrugBank ID DB09144 and CAS Number 4105-38-8, it is a small molecule chemically engineered as a highly bioavailable oral prodrug of the naturally occurring nucleoside, uridine.][1][ This structural modification significantly enhances its therapeutic utility by enabling the achievement of systemic uridine concentrations that are 4- to 6-fold higher than what is possible with equimolar doses of uridine itself.][1]
[The drug's clinical applications are governed by two distinct pharmacological mechanisms. For the treatment of hereditary orotic aciduria, a rare condition caused by a deficiency in uridine monophosphate synthase, uridine triacetate acts as a replacement therapy. Marketed as Xuriden®, it bypasses the genetic defect by providing an exogenous source of uridine, thereby restoring the pyrimidine nucleotide pool and reducing the toxic accumulation of orotic acid through feedback inhibition.][1][ In its second role, marketed as Vistogard®, it serves as a life-saving antidote for overdose or severe, early-onset toxicity resulting from the chemotherapeutic agents fluorouracil or capecitabine. In this context, it functions as a competitive antagonist, where the supplemental uridine outcompetes the toxic fluorouracil metabolite, fluorouridine triphosphate (FUTP), for incorporation into the RNA of healthy cells, thus preventing cellular damage and death.][1]
[Clinical evidence supporting its use as an antidote is robust, demonstrating a 96% survival rate in patients with fluoropyrimidine overdose, a dramatic improvement over the 16% survival rate observed in historical cohorts receiving only supportive care.][5][ The safety profile of uridine triacetate is notably favorable, characterized by generally mild and transient gastrointestinal side effects and a complete absence of listed contraindications, warnings, or precautions in its prescribing information.][7]
[Despite its proven efficacy and safety, global access to uridine triacetate is fragmented. While it has full marketing approval from the U.S. Food and Drug Administration (FDA) for both indications, its regulatory status elsewhere is varied. It recently received an orphan designation from the European Medicines Agency (EMA) for hereditary orotic aciduria but is not yet approved for marketing.][9][ In Australia, it is available only through a Special Access Scheme for emergency use.][10][ This report provides a comprehensive analysis of uridine triacetate, covering its physicochemical properties, detailed pharmacology, clinical applications, safety profile, and global regulatory landscape.]
[A complete understanding of uridine triacetate begins with its fundamental chemical and physical identity. This section provides a detailed catalogue of its nomenclature, structural properties, and key physicochemical characteristics that are foundational to its pharmaceutical function.]
[Uridine triacetate is known by a variety of names and identifiers across scientific literature, regulatory filings, and clinical practice.]
[Uridine triacetate is an acetate ester of the nucleoside uridine. Its structure is central to its function as a prodrug.]
[The chemical modification of the parent uridine molecule is a deliberate and highly effective pharmaceutical strategy. Uridine itself is a hydrophilic molecule with poor oral absorption and is subject to rapid first-pass metabolism, limiting its clinical utility when administered orally. The addition of three acetate ester groups at the 2', 3', and 5' positions of the ribose sugar renders the molecule significantly more lipophilic (fat-soluble).][15][ This increased lipophilicity enhances its ability to passively diffuse across the lipid-rich membranes of the gastrointestinal tract, leading to rapid and efficient absorption into the systemic circulation. Once absorbed, ubiquitous non-specific esterase enzymes present in the blood and tissues quickly cleave the acetate groups, releasing the active uridine moiety.][2][ This prodrug approach effectively circumvents the pharmacokinetic barriers faced by uridine, resulting in substantially higher and more reliable plasma concentrations, which is the cornerstone of its clinical efficacy.][1]
[The physical characteristics of uridine triacetate are consistent with its formulation as an oral medication.]
Table 1: Uridine Triacetate - Key Identifiers and Properties
| Property | Details |
|---|---|
| Generic Name | Uridine triacetate |
| Brand Names | Vistogard®, Xuriden® |
| DrugBank ID | DB09144 |
| CAS Number | 4105-38-8 |
| Molecular Formula | |
| Molecular Weight | Average: 370.314 g/mol; Monoisotopic: 370.101230168 g/mol |
| IUPAC Name | (2R,3R,4R,5R)-2-(acetoxymethyl)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)tetrahydrofuran-3,4-diyl diacetate |
| Key Synonyms | 2',3',5'-tri-O-acetyluridine, Vistonuridine, PN401, RG2133 |
| Appearance | White solid powder |
| Solubility | DMSO: 74-100 mg/mL; Water: 7-10 mg/mL; Ethanol: 4 mg/mL |
[The pharmacological profile of uridine triacetate is unique, as it possesses two distinct mechanisms of action that address two completely unrelated pathological conditions. The common denominator is its function as a highly efficient oral delivery system for the active moiety, uridine. Its utility is therefore defined by any condition where a systemic surplus of uridine is therapeutically beneficial.]
[In the context of hereditary orotic aciduria, uridine triacetate functions as a replacement therapy.][3][ This rare, congenital autosomal recessive disorder is caused by a defect in the bifunctional enzyme uridine monophosphate synthase (UMPS).][1][ The UMPS enzyme catalyzes the final two steps of the ][de novo][ pyrimidine biosynthetic pathway, specifically the conversion of orotic acid to uridine monophosphate (UMP), a precursor for all other pyrimidine nucleotides.][1]
[The genetic defect leads to two primary consequences: a deficiency of pyrimidine nucleotides essential for DNA and RNA synthesis, and a toxic accumulation of the precursor, orotic acid.][22][ These biochemical derangements manifest clinically as megaloblastic anemia, developmental delays, and failure to thrive.][5][ Uridine triacetate administration provides an exogenous source of uridine. This uridine can be utilized by cells through the pyrimidine salvage pathway to synthesize UMP and other necessary pyrimidine nucleotides, effectively bypassing the defective ][de novo][ pathway.][1][ This mechanism is constructive, directly replenishing a deficient metabolite. Furthermore, the restoration of normal intracellular uridine nucleotide levels re-establishes the natural feedback inhibition loop of the pyrimidine synthesis pathway, which in turn suppresses the overproduction of orotic acid and reduces its urinary excretion.][1]
[For the treatment of fluorouracil (5-FU) or capecitabine overdose, uridine triacetate acts as a competitive antidote.][1][ The anticancer drugs 5-FU and its oral prodrug capecitabine exert their cytotoxic effects through their active metabolites, which disrupt DNA and RNA synthesis in both cancerous and healthy cells.][3][ The two primary toxic metabolites are:]
[Uridine triacetate, after its conversion to uridine and subsequent phosphorylation to UTP, directly counteracts the toxicity of FUTP.][1][ By creating a massive systemic surplus of natural UTP, it effectively outcompetes the toxic FUTP for incorporation into the RNA of healthy, non-cancerous cells.][4][ This protective, antagonistic mechanism prevents RNA damage and mitigates the severe toxicities associated with fluoropyrimidine chemotherapy, such as mucositis, diarrhea, neutropenia, and cardiotoxicity.][1][ This pharmacological dichotomy showcases how a single drug, by delivering a fundamental biological molecule, can be repurposed from a constructive replacement therapy in a genetic disease to a protective antidote in iatrogenic poisoning.]
[The pharmacokinetic profile of uridine triacetate is defined by its success as a prodrug.]
[The pharmacodynamic effects of uridine triacetate differ based on the indication.]
[The clinical utility of uridine triacetate is sharply defined by its two distinct, FDA-approved indications, each supported by specific clinical evidence and marketed under a different brand name.]
[Uridine triacetate, under the brand name Xuriden®, is indicated for the treatment of hereditary orotic aciduria in both adult and pediatric patients.][1][ It is the first and only drug approved by the FDA for this ultra-rare orphan disease, which has an estimated birth prevalence of less than one in one million.][5]
[The FDA approval was based on a single-arm, open-label trial involving four patients aged 3 to 19 years.][5][ In this small cohort, treatment with uridine triacetate at 60 mg/kg once daily for six weeks, followed by a six-month extension at 60-120 mg/kg daily, demonstrated clear clinical benefit. All four patients maintained stability in their baseline hematologic parameters, including neutrophil count, white blood cell count, and mean corpuscular volume. Furthermore, three patients who were measured for growth showed either improvement or stability in weight and stable height growth over the six-month period. Notably, no adverse effects were reported during the trial.][5][ This approval highlights the regulatory flexibility and reliance on small but well-defined studies when addressing diseases with extremely small patient populations.]
[Under the brand name Vistogard®, uridine triacetate is indicated for the emergency treatment of adult and pediatric patients who experience fluoropyrimidine-related toxicity.][1][ The indication covers two specific scenarios:]
[A critical limitation of its use is that Vistogard® is not recommended for the non-emergent treatment of routine or expected adverse reactions associated with fluorouracil or capecitabine, as it may diminish the antineoplastic efficacy of these drugs.][3][ The safety and efficacy of Vistogard® when initiated more than 96 hours after the end of fluoropyrimidine administration have not been formally established by the FDA.][14]
[The FDA approval of Vistogard® was based on compelling efficacy data from two single-arm, open-label, expanded-access trials that enrolled a total of 135 patients, including both adults and children.][5][ The patient population consisted of 117 individuals treated for fluoropyrimidine overdose and 18 treated for early-onset severe toxicity.][6]
[The major efficacy endpoint was survival at 30 days or until the resumption of chemotherapy if it occurred prior to 30 days.][25][ The results were striking: the overall survival rate among patients treated with uridine triacetate was 96% (130 out of 135 patients survived).][5][ This outcome stands in stark contrast to data from a retrospective historical cohort of 25 patients who received only supportive care following a fluorouracil overdose; in that group, the survival rate was a mere 16%.][5]
[An important secondary finding from the trials was that 33% of patients with a cancer diagnosis recovered from the toxic event sufficiently to resume chemotherapy in less than 30 days.][25][ This indicates not only that the drug is life-saving but also that it enables a rapid enough recovery to allow for the continuation of essential cancer treatment.]
[The strict 96-hour window for initiating Vistogard® is a defining feature of its FDA-approved label, established based on the population studied in the pivotal trials.][14][ From a mechanistic standpoint, the protective effect of uridine should be present as long as toxic FUTP persists in cells and continues to cause damage, a timeframe which is not fully characterized and may extend beyond 96 hours. This creates a tension between the regulatory boundary and clinical reality, as some patients develop severe, life-threatening toxicities well after this window has closed.]
[Emerging evidence from case reports and small series suggests that uridine triacetate may retain efficacy when administered beyond the 96-hour mark for delayed-onset toxicities.][16][ One notable case report detailed the successful treatment of a patient who developed severe, life-threatening encephalopathy 18 days after initiating 5-FU therapy. Despite the significant delay, administration of uridine triacetate led to rapid and complete neurological recovery.][29][ While this level of evidence is insufficient for a label change, it presents a compelling option for clinicians in dire situations where no other specific antidote exists. This discrepancy highlights a critical unmet need and a priority for future research: to formally evaluate the safety and efficacy of uridine triacetate in patients with delayed-onset fluoropyrimidine toxicity.]
[The practical application of uridine triacetate is highly dependent on the specific indication, a distinction that is reinforced by its marketing under two different brand names with distinct formulations and dosing regimens. This separation is not merely a commercial strategy but a crucial safety measure designed to prevent catastrophic medication errors.]
[Uridine triacetate is available as oral granules in single-use packets, with the formulation tailored to each indication:]
[The use of distinct brand names, packet sizes, and dosing schedules represents a deliberate "human factors" engineering approach. It physically and nominally separates the treatment for the acute emergency from that of the chronic disease, minimizing the risk of a pharmacist or clinician accidentally substituting one for the other. Such an error would have severe consequences: using the smaller Xuriden® dose for an overdose would be sub-therapeutic and likely fatal, while using the high-dose Vistogard® regimen for chronic management of orotic aciduria would constitute a massive overdose.]
[The dosing for Vistogard® is designed to rapidly achieve and sustain high plasma uridine levels to counteract acute toxicity.]
[The dosing for Xuriden® is designed for chronic, once-daily maintenance therapy and is adjusted based on patient weight and clinical response.]
[Proper administration is critical for ensuring the efficacy of uridine triacetate.]
Table 2: Comparative Dosing and Administration for Vistogard® and Xuriden®
| Feature | Vistogard® | Xuriden® |
|---|---|---|
| Indication | Emergency treatment of fluorouracil or capecitabine overdose or severe early-onset toxicity 1 | Treatment of hereditary orotic aciduria 1 |
| Patient Population | Adults and pediatric patients 8 | Adults and pediatric patients 30 |
| Packet Size | 10 grams 3 | 2 grams 3 |
| Dosing Regimen | Adults: 10 g (1 packet) orallyPediatric: 6.2 g/m² orally (not to exceed 10 g/dose) 8 | Adults & Pediatric: 60 mg/kg orally, may be increased to 120 mg/kg (not to exceed 8 g/day) 30 |
| Frequency | Every 6 hours 8 | Once daily 30 |
| Total Duration | 20 doses (5 days) 8 | Chronic, long-term therapy 30 |
| Administration Instructions | Mix with 3-4 oz of soft food (applesauce, pudding, yogurt). Consume within 30 mins. Do not chew granules. Follow with 4 oz of water. Can be given via NG/G-tube.1 | Mix with 3-4 oz of soft food or small amounts of milk/infant formula. Consume immediately. Do not chew granules.30 |
| Guidance for Vomited/Missed Doses | Vomited (<2 hrs): Take another full dose immediately.Missed: Take as soon as possible. Continue with regular schedule for next dose.8 | Missed: Take as soon as possible. If near next dose, skip missed dose. Do not double dose.21 |
[The safety profile of uridine triacetate is remarkably favorable, particularly for a drug used in emergency and critical care settings. It is generally well-tolerated, with a predictable and manageable side effect profile and a notable lack of major safety concerns.]
[Data from clinical trials, primarily from the 135 patients treated with Vistogard®, show that adverse reactions are predominantly gastrointestinal in nature and are typically mild to moderate in severity.][7]
[A defining feature of uridine triacetate's safety profile is the complete absence of any listed contraindications, warnings, or precautions in the official FDA-approved prescribing information for both Vistogard® and Xuriden®.][7][ This underscores its high safety margin and suitability for emergency use where a detailed patient history may not be available. While standard precautions regarding hypersensitivity to the active ingredient or excipients apply to any medication, no specific contraindications have been identified.][22]
[Uridine triacetate has a very low potential for clinically significant drug-drug interactions.]
[The global availability of uridine triacetate is inconsistent, reflecting the complex challenges that orphan drugs and emergency antidotes face in navigating different regulatory and reimbursement landscapes. A patient's access to this life-saving medication is highly dependent on their geographic location.]
[The United States represents the most mature market for uridine triacetate, with full marketing approval for both of its indications.]
[The regulatory journey for uridine triacetate in the European Union is less advanced.]
[In Australia, uridine triacetate is not formally approved or listed on the Australian Register of Therapeutic Goods (ARTG).][38][ Access is managed through a pragmatic but non-standard pathway.]
[Based on available data, uridine triacetate has not received marketing approval in several other major jurisdictions, including Canada (Health Canada), China (NMPA), and Spain (CIMA AEMPS).][38][ This patchwork of global access underscores the significant disparities in regulatory pathways and market viability for drugs targeting ultra-rare diseases or emergency indications.]
Table 3: Global Regulatory Snapshot
| Regulatory Body/Country | Brand Name(s) | Approved/Designated Indication(s) | Approval/Access Status | Key Dates/Notes |
|---|---|---|---|---|
| USA (FDA) | Vistogard® Xuriden® | Emergency treatment of fluoropyrimidine overdose/toxicity Treatment of hereditary orotic aciduria | Approved | Vistogard® approved Dec 2015 28Xuriden® approved Sep 2015 5 |
| Europe (EMA) | Uridine triacetate | Treatment of hereditary orotic aciduria | Orphan Designation (No Marketing Authorization) | Orphan designation granted June 2024 9 |
| United Kingdom (NHS) | Uridine triacetate | Treatment of early-onset severe fluoropyrimidine toxicities | Routinely Commissioned | Policy published March 2020 40 |
| Australia (TGA) | Vistogard® | Emergency treatment of fluoropyrimidine overdose/toxicity | Special Access Scheme (SAS) (Not Registered on ARTG) | Available via centralized distribution from Peter MacCallum Cancer Centre 10 |
[Uridine triacetate stands as a significant pharmaceutical agent, defined by its dual identity as a life-saving emergency antidote and a transformative therapy for an ultra-rare genetic disease. Its development is a testament to successful rational drug design, where the chemical modification of a natural nucleoside into a lipophilic prodrug overcame critical pharmacokinetic barriers to unlock its therapeutic potential.]
[The clinical evidence for Vistogard® in treating fluoropyrimidine overdose is unequivocal, demonstrating a profound survival benefit that has established it as the standard of care for this iatrogenic emergency. For patients with hereditary orotic aciduria, Xuriden® represents the first and only targeted therapy, offering a mechanism-based treatment that addresses the underlying biochemical defect of the disease. Its safety profile is remarkably benign for a drug of such critical importance, with a lack of major toxicities, contraindications, or significant drug interactions, further solidifying its clinical value.]
[For clinical practice, this report underscores the importance of rapid diagnosis and immediate administration of Vistogard® in cases of suspected fluoropyrimidine overdose or severe early-onset toxicity. The 96-hour window for initiation is a critical parameter based on the pivotal trial data, and healthcare systems must have protocols in place to ensure timely access. For Xuriden®, the key to successful management is consistent daily administration and careful monitoring of biochemical and hematologic markers to ensure an adequate therapeutic response.]
[Looking forward, several areas warrant further investigation. The most pressing clinical question is the potential efficacy of uridine triacetate in treating delayed-onset fluoropyrimidine toxicity beyond the 96-hour window. While compelling case reports exist, formal clinical trials or well-designed registry studies are needed to provide the robust evidence required to potentially expand its approved indication. Such an expansion could save additional lives, particularly among patients with partial DPD deficiency who may present with severe toxicity later in their treatment course. Further exploration of its neuroprotective properties and its potential to mitigate toxicity from other antimetabolites that interfere with pyrimidine pathways could also unveil new therapeutic applications.]
[Finally, the fragmented global regulatory status of uridine triacetate serves as a case study in the broader challenges facing orphan drugs and emergency antidotes. The disparities in access between regions highlight the need for more harmonized and expedited regulatory pathways for drugs that, while serving small patient populations, have a transformative impact on survival and quality of life.]
Published at: October 13, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.