Small Molecule
C10H13FN2O5
163252-36-6
Viral Hepatitis B
Clevudine is a synthetic pyrimidine L-nucleoside analog developed for the treatment of chronic hepatitis B (CHB) virus infection.[1] As an L-thymidine analogue, it represented a significant therapeutic candidate due to its potent antiviral activity, which in clinical trials proved superior to the contemporary standard of care, lamivudine.[2] A defining feature of Clevudine was its uniquely sustained post-treatment virologic suppression, a characteristic attributed to its long plasma half-life and its ability to significantly reduce the intrahepatic reservoir of covalently closed circular DNA (cccDNA).[1]
Despite this profound initial promise, the clinical development of Clevudine was ultimately curtailed by a challenging long-term safety and resistance profile. While short-term studies of up to 48 weeks demonstrated excellent efficacy and a favorable safety profile, longer-term post-marketing surveillance and retrospective studies revealed two critical liabilities. First, a significant percentage of patients developed viral breakthrough and genotypic resistance after more than one year of therapy, undermining its utility for chronic treatment.[2] Second, and more decisively, long-term administration (typically beyond eight months) was associated with a significant risk of reversible, mitochondrial myopathy.[6] This dose-limiting toxicity is mechanistically linked to the drug's own metabolic pathway and its long half-life, which leads to cumulative effects on mitochondrial function in skeletal muscle.
This dual profile of promise and peril led to a divergent regulatory fate. Clevudine gained marketing approval and is used in South Korea and the Philippines.[9] However, the emergence of serious myopathy from post-marketing data in South Korea prompted the voluntary termination of its Phase III development program in the United States in 2009.[6] Clevudine's story serves as a critical case study in antiviral drug development, illustrating the importance of long-term safety and resistance monitoring. Furthermore, the scientific investigation into its mechanism of toxicity has directly informed the rational design of next-generation, liver-targeted prodrugs, such as ATI-2173, which aim to retain Clevudine's potent efficacy while mitigating its systemic toxicity.[8]
Clevudine (DrugBank ID: DB06683) is a small molecule antiviral drug classified as a synthetic pyrimidine analogue.[1] Structurally, it is an unnatural beta-L-nucleoside analog of thymidine, a key distinction from many other nucleoside analogs which possess a D-configuration.[4] It belongs to the chemical class of pyrimidine 2'-deoxyribonucleosides, which are compounds consisting of a pyrimidine base linked to a ribose sugar moiety that lacks a hydroxyl group at the 2' position.[13]
The drug is known by several synonyms and chemical names, including L-FMAU, which is an abbreviation for its full chemical name, 2'-Fluoro-5-methyl-beta-L-arabinofuranosyluracil.[1] Commercially, it has been marketed under the brand names Levovir and Revovir.[7]
The molecular formula for Clevudine is C10H13FN2O5.[13] This corresponds to an average molecular weight of 260.221 g·mol⁻¹ and a precise monoisotopic mass of 260.08084969 Da.[9] For unambiguous scientific and regulatory tracking, it is assigned the Chemical Abstracts Service (CAS) Number 163252-36-6 and the IUPAC International Chemical Identifier Key (InChI Key) GBBJCSTXCAQSSJ-XQXXSGGOSA-N.[9]
In its purified form, Clevudine is a white to off-white solid.[18] It exhibits good solubility in aqueous and organic solvents. It is reported to be soluble in water at concentrations of 30 mg/mL to 87.7 mg/mL, in dimethyl sulfoxide (DMSO) at concentrations ranging from 20 mg/mL to 100 mg/mL, in dimethylformamide (DMF) at 10 mg/mL, and in phosphate-buffered saline (PBS) at a pH of 7.2 at 5 mg/mL.[4] This solubility profile is conducive to its formulation as an oral medication and its use in laboratory research. The key chemical and physical properties of Clevudine are summarized in Table 1.
Table 1: Chemical and Physical Properties of Clevudine
Property Category | Parameter | Value | Source(s) |
---|---|---|---|
Identifiers | Generic Name | Clevudine | 13 |
DrugBank ID | DB06683 | 13 | |
CAS Number | 163252-36-6 | 13 | |
IUPAC Name | 1--5-methyl-1,2,3,4-tetrahydropyrimidine-2,4-dione | 13 | |
InChI Key | GBBJCSTXCAQSSJ-XQXXSGGOSA-N | 17 | |
Synonyms | L-FMAU; Levovir; Revovir; 2'-Fluoro-5-methyl-beta-L-arabinofuranosyluracil | 13 | |
Molecular Properties | Chemical Formula | C10H13FN2O5 | 13 |
Average Weight | 260.221 g·mol⁻¹ | 9 | |
Monoisotopic Mass | 260.08084969 Da | 13 | |
Physical Properties | Physical State | Solid | 13 |
Appearance | White to off-white | 18 | |
Water Solubility | 87.7 mg/mL | 13 | |
DMSO Solubility | 30 mg/mL - 100 mg/mL | 15 | |
Predicted Properties | logP | -0.72 (ALOGPS), -1.1 (Chemaxon) | 13 |
Hydrogen Acceptor Count | 5 | 13 | |
Hydrogen Donor Count | 3 | 13 | |
Polar Surface Area | 99.1 Ų | 13 | |
Rotatable Bond Count | 2 | 13 | |
Rule of Five Compliance | Yes | 13 |
Clevudine functions as a prodrug that must be activated within host cells to exert its antiviral effect. Following oral administration and absorption, Clevudine undergoes intracellular phosphorylation by cellular kinases to its active metabolites, clevudine monophosphate and, subsequently, clevudine 5'-triphosphate (CLV-TP).[1] This metabolic activation is predominantly mediated by cytosolic deoxycytidine kinase.[21]
The active triphosphate moiety, CLV-TP, is the direct inhibitor of the hepatitis B virus (HBV) polymerase, an enzyme that possesses reverse transcriptase activity and is essential for viral DNA replication.[14] The precise molecular interaction of CLV-TP with the HBV polymerase has been described in two distinct, and somewhat contradictory, ways. Several sources describe a classic nucleoside reverse transcriptase inhibitor (NRTI) mechanism, wherein CLV-TP competes with the natural substrate, deoxythymidine triphosphate, for incorporation into the elongating viral DNA strand. This incorporation leads to premature chain termination, thereby halting viral replication.[1]
However, a more sophisticated and distinct mechanism has also been proposed, which takes into account Clevudine's unique unnatural L-configuration. According to this model, CLV-TP acts as a non-competitive inhibitor of the HBV polymerase.[19] It binds to the enzyme's catalytic center, but due to its stereochemical conformation, it is not incorporated into the nascent viral DNA.[8] In this scenario, Clevudine does not function as a chain terminator but rather as a direct inhibitor that physically obstructs the polymerase's function.[19] This non-competitive inhibition model may better explain some of Clevudine's unique pharmacodynamic properties and differentiates it from other NRTIs like lamivudine.
A key feature that distinguishes Clevudine from many other anti-HBV agents is its significant impact on the pool of covalently closed circular DNA (cccDNA) within the nucleus of infected hepatocytes.[1] The cccDNA molecule serves as the stable transcriptional template for all viral RNAs and is the primary reason for the persistence of HBV infection and viral relapse after therapy cessation.[24] Clevudine treatment has been shown to lead to a marked reduction in intrahepatic cccDNA levels.[22] This depletion of the viral reservoir is considered the most plausible explanation for Clevudine's characteristic and clinically observed sustained post-treatment antiviral effect.[1]
Clevudine is formulated for oral administration, typically as a 30 mg capsule or tablet taken once daily.[7] Pharmacokinetic studies have demonstrated that its exposure (as measured by Cmax and AUC) is proportional to the dose administered across a range of 10 mg to 200 mg.[26]
The drug exhibits extensive distribution into tissues, with a steady-state volume of distribution in animal models that is greater than the total volume of body water, indicating significant intracellular accumulation.[21] This is consistent with its mechanism of action, which requires it to enter hepatocytes to be activated. Plasma protein binding is low, at approximately 15%, suggesting that a large fraction of the drug in circulation is free and available for tissue distribution.[21]
As a prodrug, Clevudine's primary metabolism is the intracellular phosphorylation to its active triphosphate form. The parent drug is predominantly eliminated from the body unchanged via renal excretion in the urine.[21]
A defining pharmacokinetic characteristic of Clevudine is its exceptionally long plasma half-life. Mean half-life values have been consistently reported in the range of 44 to 70 hours, with a median of 60-70 hours in some studies.[17] Due to this slow elimination, steady-state plasma concentrations are not reached until after approximately 22 days of continuous daily dosing.[21] This prolonged half-life is a critical pharmacological feature with dual, opposing consequences. On one hand, the sustained drug exposure is directly responsible for its unique and beneficial pharmacodynamic profile, particularly the durable suppression of HBV DNA that persists for months after treatment is stopped.[5] On the other hand, this same property of long-term, cumulative drug exposure is the likely driver of its primary dose-limiting toxicity, a delayed-onset mitochondrial myopathy that emerges only after chronic administration.[8] The very pharmacokinetic attribute that confers a therapeutic advantage is also intrinsically linked to its most significant safety liability.
The relationship between Clevudine dose and its antiviral effect was well-characterized in Phase II clinical trials. Pharmacodynamic modeling revealed that a once-daily dose of 30 mg achieved 97% of the maximal treatment effect, establishing this as the optimal dose for subsequent Phase III development and clinical use.[26]
The most notable pharmacodynamic property of Clevudine is its potent and sustained post-treatment antiviral activity. Multiple clinical studies have shown that after a 12-week course of therapy, significant suppression of HBV DNA levels is maintained for as long as 24 weeks (6 months) after the drug is discontinued.[2] This durable response is unique among HBV nucleoside analogs and is attributed to the drug's effect on the cccDNA reservoir.[5]
Regarding drug interactions, the primary concern documented is a potential reduction in the therapeutic efficacy of live attenuated vaccines when co-administered with Clevudine. This interaction has been noted for vaccines such as BCG, Rubella, Smallpox (Vaccinia), and Varicella zoster.[13] As Clevudine is cleared renally, caution is also advised when it is used concurrently with other nephrotoxic drugs or agents that compete for active tubular secretion, as these could potentially alter Clevudine's plasma concentrations and increase the risk of toxicity.[7] Several clinical trials have explored Clevudine in combination with other anti-HBV agents, such as adefovir and lamivudine, to assess its potential role in combination therapy regimens, particularly for treatment-experienced patients.[30]
The initial clinical evaluation of Clevudine involved Phase II dose-escalation studies designed to assess its safety, tolerability, pharmacokinetics, and antiviral activity. These trials typically evaluated once-daily oral doses ranging from 10 mg to 200 mg over treatment durations of 28 days to 12 weeks.[26] The results from these early studies were highly encouraging. Clevudine demonstrated potent, dose-dependent antiviral activity, with 12 weeks of treatment producing median reductions in serum HBV DNA of -3.2 log₁₀ copies/mL at the 10 mg dose to -4.2 log₁₀ copies/mL at the 50 mg dose.[28] Importantly, the drug was generally well-tolerated in these short-term studies, and no dose-limiting toxicities were identified, supporting its progression into larger, longer-term pivotal trials.[27]
The efficacy of Clevudine was further established in larger Phase III and Phase IV trials, where it was compared against other approved anti-HBV agents. A key 48-week, double-blind, randomized study (NCT00362635) directly compared Clevudine 30 mg daily against the then-standard-of-care, lamivudine 100 mg daily, in treatment-naïve patients with HBeAg-positive CHB.[32] This trial demonstrated the clear superiority of Clevudine. At week 48, 73% of patients in the Clevudine arm achieved undetectable serum HBV DNA levels (<300 copies/mL), compared to only 40% of patients in the lamivudine arm, a statistically significant difference (p=0.001).[3]
Other trials were conducted to compare Clevudine with adefovir dipivoxil, particularly in HBeAg-negative patients (e.g., NCT00641082).[35] A large-scale, 96-week Phase III trial (QUASH 1, NCT00496002) was initiated to compare Clevudine with adefovir in HBeAg-positive patients, but this study was terminated prematurely due to emerging safety concerns.[36] Across these studies, 24 to 48 weeks of Clevudine monotherapy consistently produced high rates of both virologic response (HBV DNA undetectable in 59% of HBeAg-positive and 92% of HBeAg-negative patients at 24 weeks) and biochemical response (alanine aminotransferase normalization in 68-86% of patients).[2] Rates of HBeAg seroconversion were observed in approximately 18% to 25% of patients after 48 weeks of treatment.[2]
Table 2: Summary of Key Clinical Trials for Clevudine in Chronic Hepatitis B
Trial Identifier | Phase | Objective | Design | Patient Population | Arms | Duration | Key Efficacy Outcomes | Status |
---|---|---|---|---|---|---|---|---|
NCT00362635 | 3 | Compare efficacy and safety of Clevudine vs. Lamivudine | Randomized, Double-Blind | HBeAg-positive, treatment-naïve | Clevudine 30 mg vs. Lamivudine 100 mg | 48 weeks | Superior HBV DNA suppression with Clevudine (73% vs. 40% undetectable) | Completed 32 |
NCT00641082 | 4 | Compare safety and antiviral activity of Clevudine vs. Adefovir | Randomized, Double-Blind | HBeAg-negative with compensated liver function | Clevudine vs. Adefovir Dipivoxil | 48 weeks | Comparison of safety and antiviral activity | Completed 35 |
NCT00496002 (QUASH 1) | 3 | Compare efficacy and safety of Clevudine vs. Adefovir | Randomized, Double-Blind, Active-Control | HBeAg-positive, treatment-naïve | Clevudine 30 mg vs. Adefovir 10 mg | Planned 96 weeks | Comparison of efficacy and safety at weeks 48 and 96 | Terminated 36 |
NCT01264133 | 4 | Evaluate Clevudine monotherapy vs. Clevudine + Adefovir combination | Randomized | Chronic Hepatitis B | Clevudine vs. Clevudine + Adefovir | N/A | Comparison of monotherapy vs. combination therapy | Terminated 30 |
NCT00798460 | 4 | Efficacy of Clevudine + Adefovir for Lamivudine resistance | Randomized, Controlled | Lamivudine-resistant CHB | Clevudine + Adefovir vs. Lamivudine + Adefovir | 12 months | Comparison of antiviral effects in resistant patients | Terminated 30 |
The compelling efficacy observed in short-term trials of up to 48 weeks did not fully translate to sustained long-term viral control. A critical disconnect emerged between the initial data, which suggested a high barrier to resistance (no resistance was detected in the 48-week trial against lamivudine), and the outcomes observed in longer-term retrospective studies.[2]
When treatment was extended beyond one year, a significant rate of virologic failure became apparent. Long-term follow-up studies of treatment-naïve patients revealed cumulative rates of viral breakthrough that increased over time, reaching 6.6% at 12 months, 22.5% at 24 months, and as high as 30.0% at 36 months.[2] This breakthrough was driven by the development of genotypic resistance. The cumulative incidence of resistance mutations also rose with treatment duration, reaching 4.6% at 12 months, 16.1% at 24 months, and 24.2% at 36 months.[2] This high rate of resistance development with prolonged monotherapy was a major liability, rendering Clevudine a less viable option for the chronic, often lifelong, treatment required for CHB.
The primary genotypic mutation associated with Clevudine resistance was identified as a substitution at codon 204 in the YMDD motif of the HBV polymerase (rtM204I).[37] This is the same mutation that confers resistance to lamivudine, indicating significant cross-resistance between the two drugs. In vitro susceptibility testing confirmed that while Clevudine-resistant mutants were also resistant to lamivudine, they generally remained susceptible to the nucleotide analogs adefovir and tenofovir, providing potential rescue therapy options.[38] Clinical risk factors independently associated with a higher likelihood of developing viral breakthrough on Clevudine therapy included being HBeAg-positive, having a high baseline serum HBV DNA level (
≥6 log10 IU/mL), and having detectable HBV DNA at week 24 of treatment.[2]
In clinical trials with treatment durations of up to one year, Clevudine was generally safe and well-tolerated.[2] The most commonly reported adverse events were mild to moderate in severity and were comparable to those seen with placebo or active comparators. These included non-specific symptoms such as fatigue, headache, dyspepsia, pruritus (itching), and abdominal pain.[2]
The most significant and dose-limiting adverse event associated with Clevudine is a drug-induced mitochondrial myopathy. This toxicity was not apparent in the initial short-term clinical trials but emerged as a serious concern from post-marketing surveillance and long-term treatment studies.[6]
The clinical presentation of Clevudine-induced myopathy is characterized by progressive proximal muscle weakness, myalgia (muscle pain), and profound fatigue.[6] Patients often report functional impairment, such as difficulty climbing stairs or rising from a seated position.[39] The diagnosis is supported by laboratory findings of significantly elevated serum creatine phosphokinase (CK) levels, typically more than twice the upper limit of normal.[6] The onset of this myopathy is notably delayed, typically appearing only after prolonged, continuous exposure to the drug, usually after at least 8 to 12 months of therapy.[2] In one long-term study, clinically significant myopathy developed in 5.9% of patients.[2] The condition was found to be reversible, with muscle strength returning and CK levels normalizing within weeks to months after Clevudine was discontinued.[2]
Histopathological examination of muscle biopsy specimens from affected patients confirms a mitochondrial etiology. The characteristic findings include the presence of "ragged-red fibers" on light microscopy, which are indicative of abnormal mitochondrial accumulation.[39] Electron microscopy reveals severe ultrastructural damage, including necrotic myofibers and extremely dysmorphic mitochondria with extensive loss or clumping of their internal cristae.[39]
The mechanism underlying Clevudine-induced myopathy is complex and distinct from that of many other NRTIs. Many NRTIs are known to cause mitochondrial toxicity by directly inhibiting the human mitochondrial DNA polymerase gamma (Pol-γ), the enzyme responsible for replicating mitochondrial DNA (mtDNA).[39] However, preclinical studies demonstrated that the active triphosphate form of Clevudine is neither a substrate for nor an inhibitor of human Pol-γ, which initially suggested a favorable mitochondrial safety profile and created a paradox when the myopathy was discovered.[21]
Further investigation revealed a more subtle, indirect mechanism. The pathophysiology is now understood to be linked to the drug's own metabolic activation pathway. The first phosphorylation step, converting Clevudine to clevudine monophosphate, is carried out by cellular kinases, including the mitochondrial enzyme thymidine kinase 2 (TK2).[8] TK2 is a critical enzyme for the maintenance of the mitochondrial nucleotide pool and is essential for normal mtDNA synthesis. The prevailing hypothesis is that chronic, long-term exposure to Clevudine acts as a constant drain on the cellular pool of TK2. This leads to a gradual "exhaustion" of the enzyme's capacity. As TK2 becomes depleted, the synthesis of mtDNA is impaired, leading to mtDNA depletion, subsequent mitochondrial respiratory chain dysfunction, and cellular energy failure in tissues with high energy demands, such as skeletal muscle. This cumulative process explains the delayed onset of the myopathy.[8] This mechanistic understanding is pivotal, as it provides a clear scientific rationale for designing next-generation prodrugs that can bypass this problematic metabolic step.
The regulatory journey of Clevudine has been geographically divergent. Based on positive results from its Phase III clinical program, Clevudine was approved for the treatment of chronic hepatitis B by the Korea Food & Drug Administration (KFDA) in 2006.[6] It is commercially available in South Korea, marketed by Bukwang Pharmaceutical under the brand names Levovir and Revovir.[9]
Following its approval in Korea, Clevudine also gained approval in the Philippines in February 2009.[9] The Japanese pharmaceutical company Eisai Co., Ltd., having licensed the development and marketing rights for Clevudine in eight Asian countries from Bukwang, launched the drug in the Philippines in 2010 under the brand name Revovir.[10]
In contrast to its status in Asia, Clevudine never received marketing approval from the U.S. Food and Drug Administration (FDA). In April 2009, Pharmasset, Inc., the company developing Clevudine for the U.S. market, announced the voluntary termination of its pivotal Phase III QUASH studies (including NCT00496002).[6]
This decision was not a direct result of adverse events observed within the U.S. trials themselves, where the reported cases of myopathy had been infrequent and mild. Instead, the termination was a proactive measure prompted by an increasing number of spontaneous post-marketing reports of more severe myopathy emerging from South Korea, where patients had been treated for much longer durations than those in the ongoing U.S. trials.[11] Pharmasset's Chief Medical Officer stated that these more severe reports from real-world use led the company to conclude that the risk-benefit ratio for Clevudine was "insufficient to continue development".[11]
This series of events provides a compelling example of the impact of global pharmacovigilance on drug development. The real-world, long-term safety data generated in one regulatory jurisdiction (South Korea) directly informed and ultimately halted the clinical development program in another (the United States). It also highlights how different regulatory agencies may arrive at different conclusions regarding a drug's risk-benefit profile; the KFDA concluded that the myopathy was infrequent and reversible, permitting continued marketing with safety warnings, whereas the emerging safety signal was deemed prohibitive for continued development in the U.S..[6]
The clinical and regulatory history of Clevudine presents a classic narrative of a drug with immense therapeutic promise ultimately limited by long-term liabilities. Its initial development was marked by significant achievements: potent antiviral activity, demonstrated superiority over the existing standard of care, and a unique pharmacodynamic profile characterized by sustained post-treatment viral suppression.[3] This latter effect, driven by its ability to reduce the stable cccDNA reservoir, positioned Clevudine as a potential breakthrough in the management of CHB.
However, Clevudine's trajectory also serves as a critical case study illustrating two fundamental challenges in the development of therapies for chronic viral infections. The first is the emergence of drug resistance with long-term use. The initially low rates of resistance observed in 48-week trials were misleading, as longer-term data revealed a cumulative incidence of viral breakthrough approaching 30% after three years, rendering it unsuitable as a long-term monotherapy.[2] The second, and more decisive, challenge was the appearance of a delayed and cumulative toxicity. The mitochondrial myopathy, not apparent in shorter pivotal trials, became the drug's defining safety issue. The development path of Clevudine underscores the indispensable value of long-term follow-up studies and robust post-marketing surveillance to fully characterize the safety and resistance profiles of drugs intended for chronic administration.
The scientific story of Clevudine did not end with the termination of its U.S. development. The detailed elucidation of its mechanism of toxicity provided a clear roadmap for rational drug design to overcome its limitations. This has led to the development of ATI-2173, a liver-targeted 5'-phosphoramidate prodrug of Clevudine.[8]
The design of ATI-2173 is a direct and elegant scientific response to the specific metabolic flaw of its parent molecule. By chemically modifying Clevudine to deliver its 5'-monophosphate form directly, ATI-2173 is engineered to bypass the first, TK2-dependent phosphorylation step that is implicated in mitochondrial toxicity.[8] This strategy is intended to achieve two primary goals: first, to mitigate or eliminate the risk of myopathy by avoiding the depletion of mitochondrial TK2 in skeletal muscle; and second, to enhance the delivery of the active moiety to the liver, its site of action, thereby potentially increasing antiviral efficacy while simultaneously reducing systemic drug exposure and its associated risks.[8] The evolution from Clevudine to ATI-2173 exemplifies the iterative nature of pharmaceutical research, where the failures and limitations of one molecule provide the crucial knowledge base needed to engineer a safer and more effective successor. Clevudine, therefore, remains a scientifically important molecule whose legacy is defined not only by its clinical shortcomings but also by the valuable lessons that are shaping the future of anti-HBV therapy.
Published at: August 28, 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.