C10H11F3N2O5
70-00-8
Metastatic Colorectal Cancer (CRC), Metastatic Gastric Cancers, Metastatic Gastroesophageal Junction Adenocarcinoma, Primary keratoconjunctivitis caused by herpes simplex virus type 2, Recurrent epithelial keratitis caused by herpes simplex 2
This report provides an exhaustive analysis of trifluridine, a fluorinated pyrimidine nucleoside with a unique dual role in modern medicine. Initially developed as an antineoplastic agent and later approved as a topical antiviral for herpetic keratitis (Viroptic), trifluridine has been successfully repurposed for systemic oncology in combination with tipiracil (Lonsurf). The monograph details its chemical properties, bifurcated pharmacology, distinct pharmacokinetic profiles, extensive clinical applications in both ophthalmology and oncology, and disparate safety considerations. Furthermore, it explores the drug's developmental history, current research landscape, and future therapeutic potential, offering a comprehensive resource for clinicians, pharmacologists, and researchers.
Trifluridine, a pyrimidine 2'-deoxyribonucleoside analogue of thymidine, has followed a remarkable and bifurcated trajectory in pharmacotherapy.[1] The molecule was first synthesized in 1964 by Heidelberger and colleagues, originally with antineoplastic intent.[3] However, early clinical investigations of intravenous trifluridine for cancer proved disappointing. The drug exhibited an exceptionally short serum half-life of approximately 12 minutes, which resulted in limited and transient therapeutic responses, halting its initial development as a systemic anticancer agent.[3]
Despite this initial setback, trifluridine found its first clinical success in a different therapeutic area. Its potent antiviral properties led to its development as a topical ophthalmic solution. In 1980, it received approval from the U.S. Food and Drug Administration (FDA) under the brand name Viroptic for the treatment of primary keratoconjunctivitis and recurrent epithelial keratitis caused by the herpes simplex virus (HSV).[5] For decades, this remained its primary clinical identity.
The story of trifluridine in oncology was revitalized by a critical scientific breakthrough in understanding its metabolism. Researchers determined that its rapid degradation and poor oral bioavailability were due to extensive first-pass metabolism by the enzyme thymidine phosphorylase (TPase).[3] This understanding led to a rational drug design strategy: combining trifluridine with tipiracil, a potent TPase inhibitor.[3] This combination, known as TAS-102 and later branded as Lonsurf, represented a "pharmacokinetic rescue" of the molecule. By preventing its degradation, tipiracil dramatically improved trifluridine's systemic exposure, allowing it to reach therapeutic concentrations when administered orally. This successful strategy culminated in the FDA approval of Lonsurf in 2015 for patients with metastatic colorectal cancer (mCRC), marking a full-circle return to its original purpose.[1] Trifluridine's journey exemplifies a powerful principle in drug development, where understanding and overcoming pharmacokinetic barriers can unlock the therapeutic potential of an otherwise ineffective molecule. This report analyzes this single chemical entity through the dual lenses of its distinct therapeutic applications, which are fundamentally defined by formulation and pharmacokinetic modulation.
A comprehensive understanding of trifluridine begins with its fundamental chemical and physical properties, which are foundational to its biological activity.
Trifluridine is identified globally through a standardized set of chemical names and registry numbers. Its systematic International Union of Pure and Applied Chemistry (IUPAC) name is 1--5-(trifluoromethyl)pyrimidine-2,4-dione.[1] It is also widely known by several common names, including trifluorothymidine (abbreviated as TFT or FTD) and α,α,α-trifluorothymidine.[5] Its unique Chemical Abstracts Service (CAS) Registry Number is 70-00-8, and its DrugBank Accession Number is DB00432.[1]
Trifluridine is an organofluorine compound classified as a pyrimidine 2'-deoxyribonucleoside.[1] Its molecular formula is
C10H11F3N2O5, and it has a molecular weight of approximately 296.20 g/mol.[5] Physically, it is a white to off-white crystalline solid.[2] Its solubility profile shows that it is freely soluble in methanol and acetone; soluble in water and ethanol; and only slightly to sparingly soluble in less polar solvents like diethyl ether and isopropyl alcohol.[5] The melting point is reported to be in the range of 189–193 °C.[11] For clinical use, the ophthalmic solution requires refrigerated storage at 2–8°C to maintain stability, while the powder form is typically stored at -20°C for research purposes.[10]
The following table provides a consolidated reference for the key chemical identifiers and properties of trifluridine.
Table 1: Key Identifiers and Chemical Properties of Trifluridine
Property | Value | Source(s) |
---|---|---|
IUPAC Name | 1--5-(trifluoromethyl)pyrimidine-2,4-dione | 1 |
Common Synonyms | Trifluorothymidine (TFT, FTD), α,α,α-trifluorothymidine, Viroptic, F3T | 2 |
CAS Number | 70-00-8 | 1 |
DrugBank ID | DB00432 | 1 |
PubChem CID | 6256 | 5 |
Molecular Formula | C10H11F3N2O5 | 5 |
Molecular Weight | 296.20 g/mol | 1 |
Canonical SMILES | C1C@@HO | |
InChIKey | VSQQQLOSPVPRAZ-RRKCRQDMSA-N | |
Physical Appearance | White to off-white crystalline powder/solid | |
Solubility Profile | Soluble in water, ethanol; freely soluble in methanol, acetone | |
Melting Point | 189–193 °C |
The pharmacological profile of trifluridine is unique, with its molecular mechanism of action being leveraged against two distinct pathologies: viral infections and cancer. While the core principle of disrupting DNA synthesis is common to both, the specific interactions and clinical strategies differ significantly.
Trifluridine's therapeutic effects are rooted in its structural similarity to the natural nucleoside thymidine, allowing it to act as an antimetabolite.
As an antiviral agent, trifluridine functions by subverting the viral replication process. It is a nucleoside analogue of deoxyuridine that, upon entering a cell, is phosphorylated by both host and viral enzymes, notably thymidine kinase, to its active triphosphate form. This activated molecule, trifluridine triphosphate, then competes with the natural substrate, thymidine triphosphate, for incorporation into the elongating viral DNA chain by viral DNA polymerase.
The critical feature of trifluridine is the trifluoromethyl (−CF3) group at the 5-position of the pyrimidine ring. This bulky, highly electronegative group prevents proper base pairing with adenine in the complementary DNA strand. This structural disruption leads to the synthesis of fragile, defective viral DNA, the production of non-functional viral proteins, an increased rate of mutation, and ultimately, the cessation of viral replication. This mechanism has demonstrated efficacy against Herpes Simplex Virus types 1 and 2 (HSV-1, HSV-2), vacciniavirus, and certain strains of adenovirus.
In oncology, trifluridine exerts its cytotoxic effects through a dual mechanism that differentiates it from other fluoropyrimidines.
First, and most importantly, is its direct incorporation into DNA. Similar to its antiviral action, trifluridine is transported into cancer cells and phosphorylated to trifluridine triphosphate. This active form is then integrated into the DNA of cancer cells, where it causes significant DNA dysfunction, induces double-strand breaks, and triggers cell death. Nonclinical studies have demonstrated a high correlation between the quantity of trifluridine incorporated into DNA and the observed cytotoxic activity, confirming this as its primary antineoplastic mechanism.
Second, trifluridine monophosphate also functions as a reversible inhibitor of thymidylate synthase (TS). TS is a critical enzyme in the de novo synthesis of pyrimidines, responsible for converting deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP), an essential precursor for DNA synthesis. This inhibitory action is analogous to that of 5-fluorouracil (5-FU), a cornerstone of chemotherapy for gastrointestinal cancers.
The dual nature of its mechanism provides a distinct advantage. The primary mechanism of action for 5-FU is TS inhibition, leading to a "thymineless death." A common mechanism of resistance to 5-FU involves the upregulation of the TS enzyme by cancer cells. While trifluridine also inhibits TS, its principal cytotoxicity derives from direct DNA incorporation. This means that even if a cancer cell has developed resistance to 5-FU by overexpressing TS, it can remain vulnerable to the DNA-damaging effects of trifluridine. This differentiated mechanism explains its clinical efficacy in patients with mCRC who have previously been treated with and are refractory to fluoropyrimidine-based chemotherapy, positioning it as a valuable therapeutic option for a chemoresistant population.
In pharmacodynamic studies, trifluridine has shown potent activity against HSV-1 and HSV-2 both in vitro and in vivo. While the potential for cross-resistance with other nucleoside analogues like idoxuridine exists, clinical evidence shows that trifluridine remains effective in treating herpetic keratitis in patients who have failed therapy with idoxuridine or vidarabine.
In the context of oncology, the combination of trifluridine/tipiracil has demonstrated significant antitumour activity in nonclinical models of both 5-FU-sensitive and 5-FU-resistant colorectal cancer cell lines. Its efficacy in 5-FU-resistant models underscores its distinct mechanism of action. Furthermore, activity has been observed in xenograft models of both KRAS wild-type and KRAS mutant human colorectal cancer, indicating its utility across different molecular subtypes of the disease.
The successful repurposing of trifluridine as a systemic oral anticancer agent is entirely dependent on its combination with tipiracil. When trifluridine is administered orally as a single agent, it is subject to rapid and extensive first-pass metabolism in the gut and liver by the enzyme thymidine phosphorylase (TPase). This enzymatic degradation converts trifluridine into an inactive metabolite, 5-(trifluoromethyl)uracil (FTY), resulting in a very short systemic half-life and negligible therapeutic concentrations.
Tipiracil is a potent and specific inhibitor of TPase. By co-formulating trifluridine with tipiracil in a fixed 1:0.5 molar ratio (the combination known as Lonsurf or TAS-102), the enzymatic degradation of trifluridine is effectively blocked. This inhibition leads to a dramatic increase in the bioavailability of trifluridine, with studies showing that its systemic exposure (as measured by the area under the curve, or AUC) is increased by as much as 37-fold compared to trifluridine administered alone. This pharmacokinetic enhancement allows orally administered trifluridine to achieve and sustain the plasma concentrations required to exert its cytotoxic effects on tumor cells.
The route of administration and formulation fundamentally dictate the pharmacokinetic (PK) behavior of trifluridine, resulting in two distinct profiles that define its separate clinical applications and safety considerations.
When administered as a 1% ophthalmic solution, trifluridine is designed for local action with minimal systemic effects.
In its oral formulation combined with tipiracil, the PK profile of trifluridine is characterized by controlled systemic exposure.
The stark contrast between these two profiles is summarized in the table below.
Table 2: Comparative Summary of Pharmacokinetic Parameters
Trifluridine's clinical utility is firmly established in two distinct fields: ophthalmology, as a topical antiviral, and oncology, as a component of a systemic oral chemotherapy agent.
As an ophthalmic solution, trifluridine (Viroptic) has been a long-standing treatment for viral eye infections.
The FDA-approved indication for trifluridine ophthalmic solution is the treatment of primary keratoconjunctivitis and recurrent epithelial keratitis caused by HSV types 1 and 2. Its clinical efficacy is well-documented; a pivotal multicenter clinical trial demonstrated a 95% response rate, defined as complete corneal re-epithelialization within a 14-day treatment period. It has also proven effective in patient populations with more challenging disease, showing a 92% response rate in individuals whose infections were unresponsive or who were intolerant to older antiviral agents like idoxuridine or vidarabine.
In the landscape of topical antivirals for herpetic keratitis, trifluridine's efficacy is comparable to that of acyclovir, with both agents being superior to older drugs such as idoxuridine. Ganciclovir has been shown to be at least as effective as acyclovir. While direct head-to-head clinical trials in humans comparing trifluridine and ganciclovir are limited, animal models have suggested equivalent healing capabilities.
The primary distinction between these agents lies in their safety and tolerability profiles. Trifluridine's mechanism of action is nonselective, as it can be phosphorylated and incorporated into the DNA of both viral and healthy host cells. This lack of selectivity is the basis for its potential for significant ocular surface toxicity, particularly with use exceeding 21 days. In contrast, ganciclovir is a prodrug that is preferentially phosphorylated by viral thymidine kinase, making it more selective for HSV-infected cells. This targeted activation results in a lower potential for toxicity to the corneal epithelium. Consequently, ganciclovir ophthalmic gel is often considered a preferred first-line topical agent by many clinicians due to its more favorable safety profile and less frequent dosing schedule.
The standard dosing regimen for trifluridine ophthalmic solution is intensive. It requires the instillation of one drop onto the affected cornea every 2 hours while awake, for a maximum of nine drops per day, until the corneal ulcer has fully re-epithelialized. Following healing, the dosage is reduced to one drop every 4 hours (a minimum of five drops per day) for an additional 7 days to prevent relapse. To mitigate the risk of ocular toxicity, continuous administration should not exceed 21 days.
Beyond its approved indication, trifluridine has important off-label uses. The U.S. Centers for Disease Control and Prevention (CDC) recommends its use for the treatment of ophthalmic infections caused by the smallpox or monkeypox (mpox) viruses. Recent preclinical research supports this, demonstrating that trifluridine is effective in vitro against a broad range of mpox isolates, including strains that are resistant to the antiviral tecovirimat. Trifluridine is also used off-label in veterinary medicine for treating ocular herpesvirus-1 infections in cats and dogs.
In combination with tipiracil as Lonsurf, trifluridine has become an important therapeutic option for patients with advanced gastrointestinal cancers.
Lonsurf is FDA-approved for the treatment of adult patients with mCRC who have been previously treated with standard chemotherapies, including a fluoropyrimidine, oxaliplatin, and irinotecan, as well as a vascular endothelial growth factor (VEGF) inhibitor. For patients whose tumors are RAS wild-type, they must have also received an epidermal growth factor receptor (EGFR) inhibitor. This places Lonsurf in the third-line or later setting for refractory disease. It is approved for use either as a single agent or in combination with bevacizumab.
Lonsurf is also approved for adult patients with metastatic gastric or GEJ adenocarcinoma who have been previously treated with at least two prior lines of chemotherapy, which must have included a fluoropyrimidine, a platinum agent, and either a taxane or irinotecan.
The approvals of Lonsurf are supported by robust data from large, randomized phase III clinical trials.
Table 3: Summary of Pivotal Clinical Trials for Lonsurf (Trifluridine/Tipiracil)
The safety profiles of trifluridine are dramatically different depending on its formulation and route of administration, reflecting the dichotomy between localized topical therapy and systemic cytotoxic treatment.
Ophthalmic Formulation (Viroptic): The adverse effects are confined to the eye and are generally mild and transient.
Oral Formulation (Lonsurf): The adverse effects are systemic and characteristic of a cytotoxic chemotherapy agent, with myelosuppression being the dose-limiting toxicity.
Table 4: Comparison of Common Adverse Effects by Formulation
Ophthalmic: Trifluridine ophthalmic solution is contraindicated in patients with a known hypersensitivity to the drug or its components. It should only be prescribed following a definitive clinical diagnosis of herpetic keratitis, as it is not effective against bacterial, fungal, or chlamydial infections.
Oral: While no absolute contraindications are listed in the prescribing information, Lonsurf carries several significant warnings and precautions :
The management of myelosuppression from Lonsurf is protocol-driven. The drug must be withheld for Grade 4 neutropenia (<500/mm³), febrile neutropenia, or platelets <50,000/mm³. Once the patient's blood counts recover to acceptable levels, treatment can be resumed at a reduced dose. The use of granulocyte-colony stimulating factors (G-CSFs) to support neutrophil counts is common, reported in 12% to 29% of patients in clinical trials.
Ophthalmic: Due to negligible systemic absorption, clinically significant systemic drug interactions are not expected. If other topical ophthalmic medications are used concurrently, they should be administered at least 5 to 10 minutes apart to avoid washout.
Oral (Lonsurf):
The research trajectory for trifluridine is heavily concentrated on expanding its role in oncology, while its application in ophthalmology remains stable but less of a focus for new development.
The clinical development of trifluridine/tipiracil (Lonsurf) is dynamic, with a large number of active trials exploring its potential beyond its current indications. The research can be categorized into several key trends.
A primary focus is the investigation of Lonsurf in novel combination regimens. Building on the success of the SUNLIGHT trial with bevacizumab, researchers are now evaluating Lonsurf in combination with other standard chemotherapies (e.g., irinotecan, oxaliplatin), targeted therapies (e.g., PARP inhibitors like talazoparib), and immunotherapies (e.g., checkpoint inhibitors like pembrolizumab and nivolumab). This strategy aims to identify synergistic effects and overcome resistance mechanisms, positioning Lonsurf not just as a late-line monotherapy but as a versatile backbone agent for combination strategies.
A second major trend is the effort to move Lonsurf into earlier stages of treatment. While currently approved for refractory disease, trials are assessing its utility in the second-line setting and even as an adjuvant or "pre-emptive" therapy for patients with minimal residual disease (as detected by circulating tumor DNA, or ctDNA) following curative-intent surgery for earlier-stage cancers.
Finally, research is underway to expand its use to new patient populations. A particularly noteworthy phase 2 trial is evaluating Lonsurf as a first-line replacement for standard fluoropyrimidines in patients with dihydropyrimidine dehydrogenase (DPD) deficiency. These patients have a genetic inability to safely metabolize 5-FU or capecitabine, and Lonsurf's different metabolic pathway presents a potentially safer and effective alternative.
While oncology research dominates, trifluridine's original identity as a broad-spectrum DNA virus inhibitor holds potential for new applications. Preclinical studies have recently demonstrated its efficacy against the mpox virus, including strains resistant to tecovirimat, the primary antiviral used for this infection. This suggests a potential for repurposing trifluridine ophthalmic solution for emerging viral threats, particularly those with ocular manifestations. However, a review of clinical trial registries indicates a lack of active trials for new ophthalmic indications, likely due to the established efficacy and superior safety profile of alternative agents like ganciclovir for its primary indication of herpetic keratitis.
The therapeutic journey of trifluridine is a compelling narrative of pharmaceutical science, demonstrating how a deep understanding of pharmacology and pharmacokinetics can transform a molecule's clinical destiny. From its initial failure as a systemic anticancer agent to its establishment as a topical antiviral, and its ultimate successful return to oncology through rational combination design, trifluridine stands as a versatile and enduring therapeutic agent.
Its clinical value is now firmly established in two disparate fields. In ophthalmology, Viroptic remains an effective treatment for herpetic keratitis, offering a reliable option that has been in use for over four decades. However, its clinical utility must be weighed against its potential for ocular surface toxicity and the availability of newer, less toxic alternatives like ganciclovir, which are often preferred for first-line topical therapy.
In oncology, Lonsurf has provided a significant and proven survival benefit for patients with heavily pre-treated, refractory metastatic colorectal and gastric cancers. The addition of bevacizumab has further solidified its role as a standard of care in the third-line setting for mCRC. This therapeutic benefit, however, comes at the cost of significant myelosuppressive toxicity, which requires diligent monitoring, patient education, and proactive management, including dose modifications and the use of supportive care agents like G-CSFs.
Looking forward, the future of trifluridine is overwhelmingly centered on its role in cancer treatment. The extensive and diverse portfolio of ongoing clinical trials suggests a strategic shift from viewing Lonsurf as a last-resort monotherapy to its potential use as a flexible backbone agent in combination regimens across multiple lines of therapy. Its oral route of administration, manageable safety profile, and distinct mechanism of action make it an attractive partner for chemotherapy, targeted agents, and immunotherapy. The exploration of its use in specialized populations, such as those with DPD deficiency, and in earlier disease settings for minimal residual disease, signals a promising expansion of its therapeutic reach, ensuring trifluridine's continued relevance in the evolving landscape of modern medicine.
Published at: September 17, 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.