Alovudine (DB06198): A Comprehensive Report on a Thymidine Analog Antiviral Agent
1. Introduction to Alovudine
Alovudine is a synthetic pyrimidine 2',3'-dideoxyribonucleoside, specifically an analog of the natural nucleoside thymidine.[1] Its primary investigation centered on its potential as an antiviral agent for the treatment of Human Immunodeficiency Virus (HIV) infection.[3] Despite demonstrating in vitro potency, particularly against certain drug-resistant HIV strains, the clinical development of Alovudine for HIV was ultimately discontinued. The cessation of its development was primarily attributed to a challenging toxicity profile observed during clinical trials and, in the later stages, a failure to meet predefined efficacy targets at doses considered tolerable.[4]
Throughout its research and development phases, Alovudine has been identified by several synonyms. These include 3'-deoxy-3'-fluorothymidine (commonly abbreviated as FLT), MIV-310 (a designation used notably during its development by Medivir), and CL 184,824.[1] A comprehensive understanding of these alternative names is essential for navigating the historical literature and tracking the compound's progression through different research and corporate entities.
The developmental trajectory of Alovudine is illustrative of the broader challenges faced in the early era of nucleoside reverse transcriptase inhibitor (NRTI) development. Many NRTIs exhibited promising antiviral activity but were ultimately hampered by significant host cell toxicity, with mitochondrial toxicity being a recurrent concern. Alovudine, despite its structural modifications such as 3'-fluorination, could not entirely escape this class-associated liability.[2] Its journey underscores the critical balance required in antiviral drug development: maximizing viral specificity and potency while minimizing detrimental off-target effects on host cellular functions. The persistent challenge lies in achieving a sufficiently wide therapeutic window, especially for chronic conditions like HIV infection that necessitate long-term treatment.
Furthermore, the history of Alovudine's development, marked by transitions between several pharmaceutical entities—from Lederle Laboratories to Medivir, then licensed to Boehringer Ingelheim, and subsequently to Presidio Pharmaceuticals—points to a compound that held recognized in vitro potential.[4] However, this potential repeatedly failed to translate into a clinically viable product, largely due to an unfavorable risk-benefit profile. Each transition in stewardship likely represented a renewed effort to overcome these inherent limitations, possibly through modified dosing strategies, different patient selection criteria, or novel combination approaches. Nevertheless, the fundamental issues of toxicity and/or insufficient efficacy at doses deemed safe appear to have remained intractable, leading to its eventual abandonment for HIV therapy. This pattern of development highlights the high-stakes, iterative, and often attritional nature of pharmaceutical research and development.
2. Chemical Profile and Properties
Alovudine's identity and characteristics are defined by its specific chemical structure and physicochemical properties.
- Nomenclature:
- Generic Name: Alovudine.[3]
- Synonyms: Key synonyms used in scientific literature and development include 3'-deoxy-3'-fluorothymidine (FLT), MIV-310, CL 184,824, CL-184824, NSC-140025, and Fluorothymidine.[1]
- Chemical Identifiers:
- DrugBank ID: DB06198.[3]
- CAS Number: 25526-93-6.[1] A deprecated CAS number, 25515-31-5, is also noted in some databases.[1]
- PubChem CID: 33039.[1]
- ChEMBL ID: CHEMBL105318.[1]
- UNII (Unique Ingredient Identifier): PG53R0DWDQ.[1]
- Structure:
- Molecular Formula: C10H13FN2O4.[1]
- Molecular Weight: 244.22 g/mol.[1]
- IUPAC Name: 1--5-methylpyrimidine-2,4-dione.[1] This nomenclature specifies the stereochemistry at the chiral centers of the fluorinated deoxyribose moiety.
- SMILES (Simplified Molecular Input Line Entry System): CC1=CN(C(=O)NC1=O)[C@H]2C[C@@H]([C@H](O2)CO)F.[1]
- InChI (International Chemical Identifier): InChI=1S/C10H13FN2O4/c1-5-3-13(10(16)12-9(5)15)8-2-6(11)7(4-14)17-8/h3,6-8,14H,2,4H2,1H3,(H,12,15,16)/t6-,7+,8+/m0/s1.[1]
- InChIKey: UXCAQJAQSWSNPQ-XLPZGREQSA-N.[1]
- Physicochemical Characteristics:
- Physical Form: Alovudine exists as a solid at room temperature.[16]
- Melting Point: Reported as 176-178 °C.[16]
- Water Solubility: Alovudine exhibits good water solubility, reported as 79.2 mg/mL by ALOGPS.[3]
- LogP (Octanol-Water Partition Coefficient): Values from various sources consistently indicate hydrophilicity: -0.6 (ALOGPS [3]), -0.23 (Chemaxon [3]), -0.3 (PubChem XLogP3 [1]), and -0.53708 (ChemScene [10]).
- pKa: The strongest acidic pKa is approximately 10.11, and the strongest basic pKa is approximately -3 (Chemaxon values [3]), indicating it is a weak acid and a very weak base.
- Polar Surface Area (PSA): Calculated as 78.87 Ų (Chemaxon [3]) or 84.32 Ų (ChemScene [10]).
- Purity: Commercially available research samples are typically offered at ≥98% purity.[10]
- Storage Recommendations: For long-term stability, Alovudine powder is best stored at -20°C. For short-term storage, 0-4°C is acceptable. Solutions, particularly in DMSO, should be stored at -20°C or -80°C for extended periods.[10]
Table 1: Summary of Key Chemical and Physical Properties of Alovudine
Property | Value | Reference(s) |
---|
Generic Name | Alovudine | 3 |
Key Synonyms | 3'-deoxy-3'-fluorothymidine (FLT), MIV-310 | 1 |
DrugBank ID | DB06198 | 3 |
CAS Number | 25526-93-6 | 1 |
Molecular Formula | C10H13FN2O4 | 1 |
Molecular Weight | 244.22 g/mol | 1 |
IUPAC Name | 1--5-methylpyrimidine-2,4-dione | 1 |
Appearance | Solid | 16 |
Melting Point | 176-178 °C | 16 |
Water Solubility | 79.2 mg/mL | 3 |
LogP (ALOGPS) | -0.6 | 3 |
pKa (Strongest Acidic) | ~10.11 | 3 |
Polar Surface Area | ~78.87 Ų (Chemaxon) | 3 |
The physicochemical profile of Alovudine, particularly its hydrophilicity (indicated by negative LogP values) and good water solubility, is characteristic of many nucleoside analogs designed for intracellular activity.[1] These properties are generally favorable for dissolution in biological fluids and interaction with cellular uptake mechanisms, such as nucleoside transporters, which are necessary for these drugs to reach their intracellular targets. Although Alovudine was primarily developed as an oral agent, its aqueous solubility would also render parenteral formulations feasible if required.
A critical structural feature of Alovudine is the fluorine atom at the 3'-position of the deoxyribose sugar. In medicinal chemistry, fluorine substitution is a widely employed strategy to modulate a drug candidate's metabolic stability, binding affinity, or electronic properties. In the context of nucleoside analogs, a 3'-fluoro modification can confer resistance to degradation by certain host or viral enzymes, such as phosphorylases, which might otherwise cleave the glycosidic bond or remove the sugar moiety. Furthermore, this substitution can alter the conformational preferences of the sugar ring (sugar pucker), which in turn can influence how the nucleoside analog interacts with the active site of target enzymes like viral reverse transcriptases or cellular DNA polymerases. This structural nuance likely contributed to Alovudine's notable in vitro potency, including its activity against some HIV strains that had developed resistance to other thymidine analogs.[2] The 3'-fluoro group, by preventing the formation of a 3'-5' phosphodiester bond, is also directly responsible for its chain-terminating mechanism of action.
3. Mechanism of Action
Alovudine exerts its biological effects through multiple mechanisms, primarily as an antiviral agent targeting HIV, but also through interactions with host cellular enzymes that contribute to its toxicity profile and have been explored for other therapeutic indications.
- Antiviral (HIV):
- Classification: Alovudine is classified as a nucleoside reverse transcriptase inhibitor (NRTI). It is a synthetic analog of the natural pyrimidine nucleoside, thymidine.[1]
- Intracellular Activation: For Alovudine to exert its antiviral effect, it must first be anabolically phosphorylated within the host cell to its active 5'-triphosphate form, Alovudine triphosphate (FLTTP). This multi-step phosphorylation is catalyzed by host cellular kinases.[2] This requirement for intracellular activation is a hallmark of NRTI drugs.
- Molecular Target in HIV: The primary molecular target of Alovudine-TP is the HIV-1 reverse transcriptase (RT) enzyme, which is essential for the conversion of the viral RNA genome into proviral DNA.[3] Alovudine-TP competes with the natural substrate, deoxythymidine triphosphate (dTTP), for incorporation into the nascent viral DNA chain.
- Mode of Action (Chain Termination): Upon incorporation into the viral DNA by HIV RT, Alovudine-TP acts as a chain terminator. The absence of a hydroxyl group at the 3'-position of its sugar moiety (due to the fluorine substitution) prevents the formation of the subsequent 3'-5' phosphodiester bond required for DNA chain elongation. This effectively halts the process of reverse transcription and, consequently, inhibits HIV replication.[2]
- Gag-Pol Polyprotein Interaction: DrugBank also lists the HIV Gag-Pol polyprotein as a target of Alovudine, with a "modulator" action.[3] The Gag-Pol polyprotein is crucial for virion assembly, maturation, and packaging of viral components. While NRTIs primarily function by inhibiting RT, indirect effects on later stages of the viral life cycle due to impaired reverse transcription are plausible. However, a direct modulatory interaction of an NRTI triphosphate with the Gag-Pol polyprotein itself is not a classical or well-established primary mechanism for this drug class and may represent a secondary effect or an area requiring further detailed investigation.
- Other Biochemical Actions (Relevant to Toxicity and Alternative Uses):
- Inhibition of Host DNA Polymerases: Beyond viral RT, Alovudine-TP can also be recognized as a substrate by host cellular DNA polymerases.
- Specific Inhibition of Mitochondrial DNA Polymerase Gamma (POLG): A critical off-target interaction of Alovudine-TP is its inhibition of human mitochondrial DNA polymerase gamma (POLG).[2] POLG is the sole DNA polymerase responsible for the replication and repair of mitochondrial DNA (mtDNA). Incorporation of Alovudine-TP into mtDNA leads to chain termination, disrupting mtDNA synthesis.
- Consequences of POLG Inhibition: The inhibition of POLG by Alovudine-TP results in the depletion of mtDNA, which encodes essential subunits of the electron transport chain complexes. This leads to impaired synthesis of these critical mitochondrial proteins, reduced oxidative phosphorylation capacity, decreased basal oxygen consumption, and diminished cellular ATP production.[2] This mitochondrial dysfunction is widely considered the molecular basis for the significant dose-limiting toxicities, such as hematological and liver toxicities, observed with Alovudine and other NRTIs that inhibit POLG. This same mechanism has also been explored for its potential anti-leukemic effects, where inducing mitochondrial dysfunction in cancer cells could be therapeutically beneficial.[2]
The dual interaction of Alovudine triphosphate with both the viral HIV RT and the host mitochondrial POLG exemplifies a common challenge in NRTI development. The therapeutic efficacy is derived from inhibiting the viral enzyme, while significant toxicity arises from inhibiting the host mitochondrial enzyme. The degree of selectivity of the NRTI triphosphate for viral RT over host POLG largely determines its therapeutic index. In the case of Alovudine, the development history suggests that this selectivity was insufficient, leading to an unacceptably narrow therapeutic window where effective antiviral doses were often associated with significant mitochondrial toxicity.
The activity of Alovudine against HIV strains harboring thymidine-associated mutations (TAMs) suggests a specific structural advantage conferred by the 3'-fluoro modification.[19] TAMs often alter the ability of RT to discriminate between natural nucleosides and NRTIs, or enhance the RT's ability to excise incorporated NRTIs. The 3'-fluoro group in Alovudine might allow its triphosphate form to be more efficiently incorporated by these mutated RTs or render the incorporated drug less susceptible to excision, thereby overcoming common resistance mechanisms. This specific activity against resistant strains was a strong rationale for its continued investigation despite early toxicity concerns.
4. Pharmacology
The pharmacological profile of Alovudine is primarily characterized by its antiviral effects, particularly against HIV, and the pharmacodynamic responses observed in clinical settings.
- Antiviral Activity (HIV):
- In Vitro Potency: Alovudine, often referred to as MIV-310 in these studies, demonstrated significant potency in inhibiting HIV replication in vitro. This activity extended to strains of HIV that had developed resistance to multiple other NRTIs.[6]
- Activity Against Resistant Strains: A key pharmacological feature of Alovudine was its effectiveness against HIV isolates harboring multiple thymidine-associated mutations (TAMs). TAMs, such as M41L, D67N, K70R, L210W, T215Y/F, and K219Q/E, are common resistance pathways selected by thymidine analogs like zidovudine (AZT) and stavudine (d4T), and they can confer cross-resistance to other NRTIs. Alovudine's ability to inhibit these TAM-containing strains was a significant driver for its development.[19] For instance, a Phase II study reported that MIV-310 (Alovudine) treatment led to a median viral load reduction of -1.60 log10 copies/mL in patients with two to three TAMs, and -1.88 log10 copies/mL in those with four or five TAMs.[19]
- Antiviral Activity (Other Viruses - Brief Mention):
- Hepatitis B Virus (HBV): In vitro studies indicated that Alovudine possesses antiviral activity against HBV.[2]
- Orthopoxvirus: Some research suggests Alovudine has activity against orthopoxviruses.[24] These findings, while indicative of a broader spectrum of DNA polymerase inhibition, were not the main focus of Alovudine's clinical development.
- Pharmacodynamic Effects in HIV Clinical Studies:
- Viral Load Reduction: Administration of Alovudine to HIV-infected individuals resulted in dose-dependent decreases in plasma HIV viral load, as measured by either HIV RNA levels or p24 antigen levels.[19]
- In a Phase II pilot study by Katlama et al. (2004), 15 highly pretreated patients receiving MIV-310 at 7.5 mg once daily for 4 weeks (added to their failing antiretroviral regimen) experienced a median viral load decrease of -1.13 log10 copies/mL. Notably, the reduction was more substantial (-1.88 log10 copies/mL) in the 11 patients not concurrently taking stavudine, compared to a -0.57 log10 copies/mL reduction in the four patients who were on stavudine.[19]
- A subsequent dose-ranging Phase II study by Ghosn et al. (2007) involving 72 treatment-experienced patients showed that Alovudine at 2 mg/day for 4 weeks resulted in a mean viral load change of -0.42 log10 copies/mL compared to placebo. The 1 mg/day dose yielded a -0.30 log10 copies/mL change versus placebo.[20]
- CD4 Cell Count: Short-term studies (e.g., 4 weeks) generally did not report significant changes in CD4 T-lymphocyte counts.[20] This is not unusual, as CD4 cell recovery often requires more prolonged viral suppression.
- Concentration-Efficacy Relationship: Early research by Flexner et al. (1994) established a relationship between Alovudine plasma concentrations (specifically, the area under the concentration-time curve over a 12-hour dosing interval, AUC12) and its antiretroviral effect. An AUC12 of 108 ng*h/mL was associated with a 50% reduction in p24 antigen levels.[25] This finding underscored the importance of achieving adequate drug exposure for antiviral activity.
- Viral Rebound: Upon discontinuation of Alovudine treatment, plasma viral load was observed to rebound in all patients in a 4-week study, indicating that, like other antiretrovirals, continuous administration would be necessary to maintain viral suppression.[19]
- Resistance Development: In the short-term (4-week) MIV-310 study by Katlama et al., no new resistance mutations in the HIV reverse transcriptase coding region were detected during the treatment period.[19] However, the potential for resistance development with long-term monotherapy or suboptimal therapy would remain a concern, as with all antiretroviral agents.
The observed antagonism or diminished efficacy of Alovudine when co-administered with stavudine (d4T) is a critical pharmacodynamic interaction.[19] Both Alovudine and stavudine are analogs of thymidine. It is highly probable that these two drugs compete for the same intracellular anabolic phosphorylation pathways, particularly for the initial phosphorylation step catalyzed by thymidine kinase, to be converted to their active triphosphate forms. Such competition could lead to reduced intracellular concentrations of the active Alovudine triphosphate, thereby diminishing its antiviral effect. Alternatively, or additionally, competition could occur at the level of HIV RT binding. This type of antagonistic interaction between NRTIs of the same natural nucleoside class has been documented and is a significant consideration in designing effective combination antiretroviral regimens.
The potent in vitro activity of Alovudine against multi-TAM HIV strains was a compelling attribute that likely fueled its continued development, especially during a period when treatment options for drug-resistant HIV were limited and the need for novel agents was acute.[19] However, the translation of this in vitro promise to robust, durable clinical efficacy at tolerable doses proved challenging. The modest viral load reductions observed in larger clinical trials at the lower, safer doses (e.g., -0.42 log10 copies/mL for the 2mg daily dose [20]) may have been insufficient to provide long-term virologic control or to be competitive with other emerging antiretroviral agents. This disparity between high in vitro potency against resistant virus and the actual clinical utility achievable within the constraints of patient safety highlights a common and significant hurdle in drug development. Achieving adequate intracellular concentrations of Alovudine triphosphate to effectively inhibit highly resistant viral variants, without concomitantly causing unacceptable levels of mitochondrial toxicity, appears to have been the central, unresolved challenge.
5. Pharmacokinetics in Humans (derived from HIV clinical trials)
The pharmacokinetic (PK) profile of Alovudine in humans was investigated in several early-phase clinical trials, primarily focusing on oral administration.
- Administration Route: Alovudine was predominantly developed and studied as an oral formulation for the treatment of HIV infection.[7] Oral delivery offers significant advantages in terms of patient convenience and adherence for chronic therapies like HIV management.
- Absorption and Bioavailability:
- Alovudine is absorbed following oral administration in humans.[25]
- A dedicated clinical trial (NCT00002260), sponsored by Lederle Laboratories, was conducted to specifically assess the effect of food on the oral bioavailability and pharmacokinetic profile of Alovudine (FLT) in asymptomatic HIV-infected individuals.[13] The detailed outcomes of this food-effect study are not available in the provided research snippets, but its execution indicates that food interactions were a recognized consideration in its development.
- Distribution:
- Specific data on the tissue distribution of Alovudine in humans (e.g., volume of distribution, penetration into specific tissues or viral sanctuary sites like the central nervous system or genital tract) are limited in the provided snippets.
- Animal studies conducted in rats, utilizing microdialysis techniques, indicated that Alovudine distributes into blood, brain, and muscle tissue.[27] While animal data can be suggestive, direct extrapolation to human tissue distribution requires caution. For an antiretroviral agent, adequate penetration into viral reservoirs is a critical factor for long-term efficacy.
- Metabolism:
- The key metabolic process essential for Alovudine's antiviral activity is its intracellular phosphorylation to the active 5'-triphosphate derivative (Alovudine-TP or FLTTP). This anabolic conversion is carried out by host cellular kinases.[2]
- Comprehensive information regarding other metabolic pathways, such as hepatic metabolism (e.g., involvement of cytochrome P450 enzymes) or the formation and clearance of inactive catabolites, is not detailed in the human pharmacokinetic data available in the provided snippets.
- Elimination:
- The route and mechanisms of Alovudine elimination from the human body are not extensively detailed in the snippets. It is mentioned that Alovudine is cleared from the body in a manner similar to that of zidovudine (AZT).[22] AZT is primarily eliminated via hepatic glucuronidation to an inactive metabolite, followed by renal excretion of both the parent drug and the glucuronide. If Alovudine follows a similar pattern, renal function could influence its clearance.
- Key Pharmacokinetic Parameters from Human Studies (Flexner et al., 1994; J Infect Dis. 1994 Dec;170(6):1394-403 [25]):
- Early clinical development involved concentration-controlled trials to define the relationship between drug exposure and both antiviral activity and toxicity.
- Area Under the Curve (AUC): The AUC over a 12-hour dosing interval (AUC12) was a key PK parameter monitored. Target AUC12 values of 50, 100, or 200 ng*h/mL were evaluated for efficacy.
- An AUC12 of 108 ng*h/mL was found to correlate with a 50% reduction in HIV p24 antigen levels, providing an early estimate of an effective exposure level.
- A critical finding was that an AUC12 ≥ 300 ng*h/mL was associated with unacceptable hematologic toxicity, establishing an exposure threshold for safety concerns.
- The plasma half-life (t1/2) of Alovudine is not explicitly stated in these summaries but would be a standard parameter determined in comprehensive PK analyses.
- Drug Interactions Affecting Pharmacokinetics/Pharmacodynamics:
- Stavudine: Co-administration with stavudine, another thymidine analog NRTI, appeared to reduce the antiviral efficacy of Alovudine.[19] This suggests a potential pharmacokinetic interaction (e.g., competition for intracellular phosphorylation pathways) or a pharmacodynamic interaction at the level of HIV RT.
- Live Vaccines: Alovudine may decrease the therapeutic efficacy of various live attenuated vaccines (e.g., Adenovirus type 7, Anthrax, BCG, Chikungunya vaccine).[3] This is likely due to the antiproliferative effects of Alovudine on rapidly dividing immune cells necessary for an effective vaccine response.
- Probenecid and Quinidine (Animal Data): Studies in rats suggested that probenecid and quinidine could influence the transport of Alovudine into the brain.[27] The clinical relevance of this finding for human CNS pharmacokinetics was not established in the provided data.
The pharmacokinetic data, particularly the early findings from Flexner et al. [25], revealed a critical characteristic of Alovudine: a narrow therapeutic window. The plasma exposure levels associated with antiviral efficacy (AUC12 ~100 ng*h/mL) were uncomfortably close to those associated with dose-limiting hematologic toxicity (AUC12 ≥ 300 ng*h/mL). This relatively small margin (less than threefold) between effective and toxic concentrations would have presented substantial difficulties in achieving consistent and safe dosing across a diverse patient population, given the inherent inter-individual variability in drug absorption, distribution, and metabolism. This narrow therapeutic index likely became a persistent challenge throughout its development and was a significant factor contributing to its eventual discontinuation for HIV treatment.
Furthermore, the absence of detailed information in the provided human data concerning Alovudine's broader systemic metabolism (beyond intracellular phosphorylation) and its specific excretion pathways represents a notable gap. Without a comprehensive understanding of these ADME (Absorption, Distribution, Metabolism, Excretion) properties, predicting potential drug-drug interactions mediated by metabolic enzymes (e.g., cytochrome P450 system) or drug transporters, and determining appropriate dose adjustments for individuals with renal or hepatic impairment, would be difficult. Such information is crucial for the safe co-administration of antiretrovirals, which are often part of complex polypharmacy regimens in HIV management, and for ensuring safety in special patient populations. The limited availability of these data in the reviewed material might suggest that either these comprehensive ADME studies were not extensively conducted or their results were not widely published, possibly because Alovudine's development was halted before these later-stage characterizations were completed or deemed necessary.
6. Clinical Development for HIV Infection
The clinical development of Alovudine for HIV infection spanned over a decade and involved several pharmaceutical entities, reflecting both its perceived therapeutic potential against resistant viral strains and the significant challenges encountered in translating this potential into a safe and effective medicine.
- Historical Overview:
Alovudine's journey as an anti-HIV agent began in the early 1990s. Initial investigations and early-phase clinical trials were conducted by Lederle Laboratories.13 Subsequently, Medivir AB took on its development, referring to the compound as MIV-310, and advanced it through Phase IIa studies.5 In July 2003, Medivir out-licensed MIV-310 to Boehringer Ingelheim, a major pharmaceutical company, which continued its clinical evaluation.4 However, Boehringer Ingelheim discontinued the development of Alovudine in March 2005.4 Following this, in December 2006, Medivir licensed Alovudine (MIV-310) to Presidio Pharmaceuticals, a company focused on antiviral drug development.6 The ultimate fate of Alovudine under Presidio's stewardship is not clearly detailed in the provided materials.
- Key Clinical Trials:
A summary of significant clinical trials involving Alovudine for HIV infection is presented in Table 2.
Table 2: Overview of Significant Alovudine Clinical Trials in HIV
NCT ID / Study ID | Phase | Sponsor(s) | Patient Population | Key Dosing Regimen(s) | Primary Endpoint(s) | Key Efficacy Outcome (Viral Load Δ) | Key Safety/Toxicity Findings | Status & Reason for Discontinuation (if specified) | Reference(s) |
---|
NCT00002254 | Not Applicable (Early Phase) | Lederle Laboratories | AIDS or AIDS-Related Complex (ARC) patients | Ascending multiple oral doses | Safety, Tolerance, PK | N/A in snippet | N/A in snippet | Completed | 13 |
NCT00002271 | Not Applicable (Early Phase) | Lederle Laboratories | Asymptomatic HIV-infected subjects | Ascending single oral doses | PK | N/A in snippet | N/A in snippet | Completed | 13 |
NCT00002260 | Not Applicable (Early Phase) | Lederle Laboratories | Asymptomatic HIV-infected subjects | Oral, crossover design | Effect of food on bioavailability & PK | N/A in snippet | N/A in snippet | Completed | 13 |
Flexner et al. 1994 (PMID: 7995977) | Phase I/II (CCTs) | NIAID AIDS Clinical Trials Group (ACTG) | HIV-infected subjects | Concentration-controlled oral doses targeting specific AUC12 values | p24 antigen change, safety | AUC12 108 ng*h/mL: 50% p24 reduction | Hematologic toxicity at AUC12 ≥ 300 ng*h/mL | Completed; established PK/PD/Toxicity link | 25 |
Katlama et al. 2004 (PMID: 15362662) | Phase IIa (Pilot) | Medivir (MIV-310) | 15 highly pretreated patients, failing ART, ≥2 TAMs | 7.5 mg MIV-310 PO QD for 4 weeks (add-on) | Plasma viral load reduction at week 4 | Median VL Δ: -1.13 log10 (-1.88 log10 if no stavudine) | Well tolerated; no SAEs; no treatment withdrawals | Completed | 19 |
Ghosn et al. 2007 (PMID: 17461857) | Phase II | Boehringer Ingelheim | 72 antiretroviral-experienced patients, NRTI-resistant HIV (≥2 TAMs) | 0.5 mg, 1 mg, or 2 mg Alovudine PO QD vs. placebo for 4 weeks (add-on) | Mean viral load reduction (baseline to week 4) | 2 mg: -0.42 log10 vs. placebo; 1 mg: -0.30 log10 vs. placebo | Well tolerated at these low doses | Completed | 20 |
NCT02232581 | Phase II | Boehringer Ingelheim | Nucleoside-experienced HIV-infected subjects experiencing virologic failure | N/A in snippet | Antiviral activity, Safety | N/A in snippet (but likely led to discontinuation decision) | N/A in snippet | Completed; Development stopped by BI due to not meeting target efficacy 4 | 13 |
*N/A: Not Available in provided snippets.*
- Efficacy Findings Summary:
Alovudine consistently demonstrated antiviral activity in human clinical trials, evidenced by reductions in HIV viral load. This activity was particularly noted in treatment-experienced patient populations harboring HIV strains with resistance to other NRTIs, including those with multiple TAMs.19
The magnitude of viral load reduction appeared to be dose-dependent. For instance, a 7.5 mg daily dose of MIV-310 (Alovudine) in a small pilot study yielded a median viral load decrease of -1.13 log10 copies/mL after 4 weeks, with a more pronounced effect (-1.88 log10) observed in patients not concurrently taking stavudine.19 However, at lower, presumably safer doses (e.g., 2 mg daily), the mean viral load reduction was more modest, around -0.42 log10 copies/mL compared to placebo after 4 weeks.20
Importantly, in short-term studies, cessation of Alovudine treatment led to a rebound in viral load, underscoring the necessity for continuous therapy for sustained viral suppression.19 During one 4-week treatment course with MIV-310, no new resistance mutations in the HIV RT gene were detected, though longer-term studies would be needed to fully assess its resistance profile.19
- Drug Interactions Noted in Clinical Context:
- Stavudine: A significant clinical observation was the reduced antiviral efficacy of Alovudine when co-administered with stavudine.[19]
- Live Vaccines: As a compound affecting DNA synthesis, Alovudine was anticipated to potentially reduce the efficacy of live vaccines.[3]
The clinical development pathway of Alovudine reveals a strategic shift in dosing over time, largely driven by early emerging toxicity concerns. Initial studies, such as those by Flexner et al. [25], likely explored higher drug exposures which led to the identification of dose-limiting hematologic toxicities. Consequently, subsequent Phase II trials conducted by Medivir (using 7.5 mg of MIV-310 [19]) and particularly by Boehringer Ingelheim (testing doses of 0.5 mg, 1 mg, and 2 mg of Alovudine [23]) focused on significantly lower doses. This dose de-escalation strategy was a clear attempt to navigate away from the identified toxicities and find a safer therapeutic window. However, this approach appears to have compromised the drug's antiviral potency.
The decision by Boehringer Ingelheim in March 2005 to halt Alovudine's development because it "did not achieve the target level of efficacy which had previously been defined" [4] is a pivotal point. This suggests that at the lower, more tolerable doses, the magnitude of viral load reduction was not considered sufficiently robust or clinically meaningful to warrant further investment, especially in the context of an evolving HIV treatment landscape where more potent and safer antiretroviral agents were becoming available. This outcome implies that the therapeutic window for Alovudine was indeed too narrow: doses low enough to mitigate serious toxicity concerns were not high enough to produce a compelling and durable antiviral effect. The focus on treatment-experienced patients with resistant virus, while addressing a high unmet need, also presents a challenging population for demonstrating substantial efficacy due to complex pre-existing resistance patterns and the potential for cumulative drug toxicities from prior regimens.
7. Safety, Tolerability, and Toxicity
The clinical utility of Alovudine was significantly undermined by its safety and toxicity profile, which became apparent during its development.
- Adverse Event Profile from Clinical Trials:
While short-term administration of Alovudine at lower doses (e.g., 0.5 mg to 2 mg daily for 4 weeks, or 7.5 mg daily for 4 weeks in a small pilot study) was reported as generally well-tolerated in some studies 19, a consistent theme throughout its development was the emergence of dose-dependent safety concerns that ultimately led to the cessation of its initial development programs.20
- Dose-Limiting Toxicities (DLTs):
The primary dose-limiting toxicities associated with Alovudine were hematological and hepatic.
- Hematological Toxicity / Bone Marrow Suppression: This was identified early as a primary target organ of toxicity.[4] Manifestations included anemia and leukopenia.[7] The concentration-controlled trials by Flexner et al. (1994) specifically linked unacceptable hematologic toxicity to Alovudine plasma exposures where the AUC12 was ≥ 300 ng*h/mL.[25] This bone marrow suppression is a prominent and well-documented toxicity of FLT.[22]
- Hepatotoxicity / Liver Toxicity: Liver toxicity was also a significant concern observed in clinical trials.[7] Reports mentioned side effects such as hepatic failure and hyperlactatemia, which are consistent with underlying mitochondrial dysfunction.[7]
- Peripheral Neuropathy: While less emphasized in the primary HIV trial snippets, peripheral neuropathy was noted as a toxicity in therapeutic FLT trials in a broader context.[31]
- Underlying Mechanism of Toxicity:
The principal mechanism underlying Alovudine's significant toxicities is its off-target inhibition of human mitochondrial DNA polymerase gamma (POLG).2 Alovudine triphosphate, the active form of the drug, is incorporated into mitochondrial DNA (mtDNA) by POLG, leading to chain termination and subsequent mtDNA depletion. This impairment of mtDNA replication and maintenance disrupts the synthesis of essential mitochondrially-encoded proteins that are critical components of the oxidative phosphorylation system. The resulting mitochondrial dysfunction manifests as reduced ATP production, impaired cellular respiration, and can lead to the observed organ toxicities, particularly in tissues with high energy demand or rapid cell turnover, such as the bone marrow and liver.7 Side effects like anemia, hepatic failure, and hyperlactatemia are classical manifestations of NRTI-induced mitochondrial toxicity.
Table 3: Summary of Clinically Observed Adverse Events and Dose-Limiting Toxicities for Alovudine in HIV Treatment
Adverse Event/Toxicity | System Organ Class | Severity/Nature | Associated Dose/Exposure (if known) | Frequency/Incidence (if known) | Reference(s) |
---|
Hematologic Toxicity (general) | Blood and lymphatic system disorders | Dose-limiting; Unacceptable | AUC12 ≥ 300 ng*h/mL | Not specified | 25 |
Bone Marrow Suppression | Blood and lymphatic system disorders | Primary target organ of toxicity; Severe | Higher therapeutic doses | Not specified | 4 |
Anemia | Blood and lymphatic system disorders | Symptom of mitochondrial toxicity/bone marrow suppression | Therapeutic doses | Not specified | 7 |
Leukopenia | Blood and lymphatic system disorders | Component of hematologic toxicity | Therapeutic doses | Not specified | 7 |
Liver Toxicity / Hepatotoxicity | Hepatobiliary disorders | Dose-limiting; Severe | Higher therapeutic doses | Not specified | 7 |
Hepatic Failure | Hepatobiliary disorders | Side effect compatible with mitochondrial mechanism | Therapeutic doses | Not specified | 7 |
Hyperlactatemia | Metabolism and nutrition disorders | Side effect compatible with mitochondrial mechanism | Therapeutic doses | Not specified | 7 |
Peripheral Neuropathy | Nervous system disorders | Noted in FLT therapeutic trials | Therapeutic doses | Not specified | 31 |
The pattern of dose-limiting toxicities observed with Alovudine—notably hematological suppression and hepatotoxicity, both strongly linked to mitochondrial dysfunction via POLG inhibition—is characteristic of several older dideoxynucleoside NRTIs, such as zalcitabine (ddC), didanosine (ddI), and stavudine (d4T). Despite being a fluorinated thymidine analog, a structural modification often intended to improve safety or potency, Alovudine ultimately shared this critical class-specific toxicity. This suggests that the structural features responsible for its interaction with HIV RT also permitted significant interaction with host POLG, leading to an unfavorable safety profile for long-term administration as required for HIV treatment.
The development of Fosalvudine tidoxil, a prodrug of Alovudine (FLT), represented an attempt to potentially mitigate the parent drug's toxicity or improve its pharmacokinetic profile.[7] However, preclinical studies with Fosalvudine tidoxil also demonstrated evidence of mitochondrial hepatotoxicity in animal models, including mtDNA depletion in the liver.[7] This finding suggests that the inherent mitochondrial liability of the active FLT moiety was difficult to overcome through a simple prodrug strategy, as the active compound released intracellularly would still exert its off-target effects on POLG.
8. Discontinuation of Development for HIV
The clinical development of Alovudine for the treatment of HIV infection was ultimately discontinued prior to regulatory approval, following a complex history involving multiple pharmaceutical developers and evolving assessments of its risk-benefit profile.
- Chronology and Key Decisions:
- Initial development efforts faced setbacks due to dose-dependent safety concerns, particularly hematological and liver toxicities, which led to an early halt in some development programs.[20]
- Boehringer Ingelheim, after licensing MIV-310 (Alovudine) from Medivir, conducted further clinical trials. However, in March 2005, Boehringer Ingelheim announced the discontinuation of Alovudine's development. The primary reason cited was that while the drug demonstrated antiviral activity, it "did not achieve the target level of efficacy which had previously been defined" in their clinical trial program (likely NCT02232581).[4]
- Some reports also attribute the discontinuation to its "limited effect on multi-drug resistant virus" in a clinical context.[6] This contrasts somewhat with other data highlighting its in vitro potency against such strains, suggesting a potential disconnect between in vitro activity and clinically achievable, safe efficacy.
- Following Boehringer Ingelheim's decision, Medivir AB out-licensed Alovudine (MIV-310) to Presidio Pharmaceuticals in December 2006.[6] The specific development activities and ultimate fate of Alovudine under Presidio Pharmaceuticals are not clearly detailed in the provided snippets, but it did not lead to market approval for HIV.
- Primary Reasons for Discontinuation for HIV Treatment:
- Significant and Dose-Limiting Toxicity Profile: The most critical factor was Alovudine's association with serious adverse effects, primarily bone marrow suppression (leading to hematological toxicities like anemia and leukopenia) and hepatotoxicity. These toxicities were dose-dependent and linked to its inhibition of mitochondrial DNA polymerase gamma.[4]
- Insufficient Clinical Efficacy at Tolerable Doses: While Alovudine showed antiviral activity, particularly against some resistant HIV strains, it failed to meet predefined efficacy endpoints in later-stage clinical trials conducted by Boehringer Ingelheim, especially at doses that were considered to have a more manageable safety profile.[4] The modest viral load reductions observed in some Phase II trials (e.g., approximately -0.42 log10 copies/mL with 2 mg/day [20]) might have been deemed insufficient for durable virologic suppression or uncompetitive with other available or emerging antiretroviral therapies.
- Unfavorable Risk-Benefit Assessment: The combination of a significant toxicity profile and modest or insufficiently robust clinical efficacy at safer doses ultimately led to an unfavorable risk-benefit assessment for long-term HIV treatment.
- Lack of Clear Therapeutic Advantage: Given its toxicity concerns, Alovudine did not demonstrate a clear overall therapeutic advantage over existing NRTIs or other antiretroviral classes that were becoming available during its development period.[4]
The decision by Boehringer Ingelheim, a major pharmaceutical company, to discontinue Alovudine's development in 2005 was a significant turning point.[4] This decision, explicitly based on the failure to meet target efficacy levels, strongly suggests that even if the toxicity could be somewhat managed by using lower doses, the resultant antiviral effect was not compelling enough to warrant further investment. The HIV treatment landscape was rapidly evolving, with newer agents offering improved potency, safety, and convenience, setting a higher bar for new entrants.
The subsequent out-licensing of Alovudine to Presidio Pharmaceuticals in late 2006 indicated that some potential was still perceived in the compound, perhaps for niche applications, specific patient populations, or in combination with other novel agents.[6] Smaller pharmaceutical companies sometimes undertake the development of higher-risk assets that larger organizations de-prioritize. However, the absence of prominent subsequent development news or regulatory filings from Presidio for Alovudine in the HIV space (within the scope of the provided information) suggests that this later effort also likely did not overcome the fundamental challenges associated with the drug.
There appears to be a nuanced, and at times seemingly contradictory, narrative regarding Alovudine's efficacy against multi-drug resistant (MDR) HIV. While several sources highlight its potent in vitro activity against resistant strains, including those with multiple TAMs [19], other reports cite its "limited effect on multi-drug resistant virus" as a reason for discontinuation.[6] This apparent discrepancy might be reconciled by considering the difference between in vitro potency and clinically achievable efficacy at tolerable doses. It is plausible that Alovudine was indeed active against certain MDR strains in laboratory settings, but achieving the necessary plasma and intracellular concentrations to replicate this effect robustly in patients, without inducing unacceptable toxicity, proved to be the insurmountable hurdle. The term "limited effect" in the clinical context could therefore refer to an insufficient magnitude or durability of viral suppression in patients with MDR-HIV when Alovudine was administered at doses constrained by its safety profile.
9. Development and Regulatory History
Alovudine's path through the pharmaceutical development pipeline was characterized by the involvement of multiple entities over more than a decade.
- Key Developers:
- Lederle Laboratories (later part of Wyeth): Involved in the initial development and early Phase I pharmacokinetic and safety studies of Alovudine (then often referred to as FLT) in the early to mid-1990s. Trials such as NCT00002254, NCT00002271, and NCT00002260 were sponsored by Lederle.[13]
- Medivir AB: This Swedish pharmaceutical company further developed Alovudine, designating it MIV-310. Medivir conducted Phase IIa clinical trials, with results reported around 2002-2004, demonstrating antiviral activity, particularly against resistant HIV strains.[5]
- Boehringer Ingelheim: In July 2003, Boehringer Ingelheim licensed MIV-310 (Alovudine) from Medivir for further clinical development. They conducted additional clinical trials, including dose-ranging Phase II studies. However, Boehringer Ingelheim terminated the development of Alovudine in March 2005, citing that the drug did not meet predefined efficacy targets.[4]
- Presidio Pharmaceuticals, Inc.: Following Boehringer Ingelheim's discontinuation, Medivir out-licensed Alovudine (MIV-310) to Presidio Pharmaceuticals in December 2006.[6] Presidio was a company focused on developing therapeutics for viral infections and liver diseases. The extent and outcome of Presidio's development efforts with Alovudine are not clearly detailed in the provided information beyond the licensing agreement.
- Regulatory Status:
- Alovudine did not receive marketing approval from major regulatory agencies like the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA) for the treatment of HIV infection. Its development was discontinued before reaching the stage of a New Drug Application (NDA) or Marketing Authorisation Application (MAA) submission for this indication.
- Clinical trial authorizations (e.g., Investigational New Drug applications or equivalents) were presumably obtained in the respective regions where trials were conducted (USA, Europe).
The development history of Alovudine serves as a compelling case study illustrating the high attrition rates and complexities inherent in pharmaceutical R&D, particularly for antiviral agents intended for chronic use. The journey of Alovudine, involving multiple handoffs between different pharmaceutical companies, reflects a common pattern where a compound with initial scientific promise faces significant hurdles in clinical translation. Larger companies may de-prioritize such assets if the risk-benefit profile or commercial viability does not meet their stringent criteria, sometimes leading to opportunities for smaller, more specialized companies to acquire and attempt to salvage these programs, perhaps by exploring niche indications, alternative formulations, or novel combination strategies. In Alovudine's case, despite these continued efforts, the fundamental challenges related to its therapeutic index for HIV treatment appear to have remained unresolved.
10. Alternative Investigational Avenues (Brief Mention)
While Alovudine's development as an oral antiretroviral for HIV was discontinued, its unique biochemical properties led to its exploration in other, distinct medical applications.
- Radiolabeled 18F-FLT for PET Imaging:
One of the notable alternative applications of Alovudine (as FLT) is its use as a positron emission tomography (PET) tracer when labeled with fluorine-18 (18F−FLT). In this context, 18F−FLT serves as a marker of cellular proliferation. Thymidine is taken up by proliferating cells and phosphorylated by thymidine kinase 1 (TK1), an enzyme whose activity is closely linked to the S-phase of the cell cycle and thus to DNA synthesis. 18F−FLT mimics thymidine in this uptake and initial phosphorylation process. The resulting 18F−FLT-monophosphate becomes metabolically trapped within the cell, allowing for the visualization and quantification of cellular proliferation rates in vivo using PET imaging.22
This diagnostic application is fundamentally different from its therapeutic use. For PET imaging, 18F−FLT is administered in microdoses (typically micrograms), which are pharmacologically inactive and orders of magnitude lower than the milligram doses required for therapeutic effect. Consequently, the toxicities associated with therapeutic doses of Alovudine are not a concern with 18F−FLT PET imaging.22 18F−FLT PET has been investigated as a tool for oncological imaging, including assessing tumor proliferation, monitoring response to cancer therapy, and potentially differentiating tumors from inflammatory processes, where it is sometimes considered less susceptible to inflammatory uptake than the more commonly used PET tracer 18F-fluorodeoxyglucose (18F−FDG).18
- Anti-leukemic Potential:
The very mechanism that contributed to Alovudine's toxicity in HIV patients—its inhibition of mitochondrial DNA polymerase gamma (POLG)—became the rationale for investigating its potential as an anti-cancer agent, particularly for acute myeloid leukemia (AML). Cancer cells, especially those with high metabolic demands or specific dependencies on mitochondrial function, might be more susceptible to POLG inhibition than normal cells. Studies have shown that Alovudine can deplete mitochondrial DNA, reduce mitochondrial protein synthesis, decrease oxidative phosphorylation, and consequently inhibit cell proliferation and viability in AML cells. Furthermore, Alovudine was observed to promote monocytic differentiation in AML cells.2 Systemic administration of Alovudine in animal xenograft models of AML demonstrated a reduction in leukemic growth.12 This avenue of research represents an attempt to repurpose a compound by leveraging a previously undesirable off-target effect (mitochondrial toxicity) as a potentially selective therapeutic mechanism in a different disease context (cancer).
The successful application of 18F−FLT as a PET imaging agent for cellular proliferation is a prime example of how a compound that fails as a systemic therapeutic due to toxicity at therapeutic doses can still find valuable utility. The core biochemical interaction of FLT with thymidine kinase, leading to intracellular trapping, is exploited in a diagnostic setting where the administered doses are too low to elicit pharmacological or toxicological effects. This repurposing highlights the importance of understanding a molecule's fundamental biological interactions, which can sometimes be harnessed for entirely different applications than initially envisioned.
Similarly, the exploration of Alovudine for AML by targeting its known mitochondrial effects represents a strategic effort to turn what was a liability in HIV treatment (mitochondrial toxicity) into a potential therapeutic advantage in oncology. The hypothesis is that certain cancer cells might exhibit a greater reliance on mitochondrial function or possess a heightened vulnerability to mitochondrial damage compared to normal tissues, thereby creating a potential therapeutic window for POLG inhibitors like Alovudine. This approach is a common theme in drug repositioning, where understanding the detailed mechanisms of action and off-target effects can open new avenues for previously shelved compounds.
11. Comprehensive Conclusion
Alovudine (3'-deoxy-3'-fluorothymidine, FLT, MIV-310), a synthetic thymidine analog, was primarily developed as a nucleoside reverse transcriptase inhibitor for the treatment of HIV infection. Its mechanism of action involves intracellular phosphorylation to Alovudine triphosphate, which then inhibits HIV reverse transcriptase and causes viral DNA chain termination. Alovudine demonstrated notable in vitro potency, particularly against HIV strains resistant to other thymidine analogs due to thymidine-associated mutations (TAMs), which was a significant driver for its clinical investigation during a period of pressing need for new antiretrovirals.
However, the clinical development of Alovudine for HIV was ultimately unsuccessful and discontinued. The primary reasons for its cessation were a challenging safety and toxicity profile, characterized by dose-limiting hematological (bone marrow) suppression and hepatotoxicity. These adverse effects were mechanistically linked to the off-target inhibition of host mitochondrial DNA polymerase gamma by Alovudine triphosphate, leading to mitochondrial dysfunction. While lower doses of Alovudine were better tolerated, they generally failed to achieve a sufficiently robust or durable antiviral efficacy to meet the evolving standards of HIV care or to offer a clear therapeutic advantage over other available agents. The narrow therapeutic window between effective antiviral concentrations and toxic concentrations proved to be a critical, insurmountable hurdle.
The journey of Alovudine through multiple pharmaceutical developers—Lederle Laboratories, Medivir, Boehringer Ingelheim, and Presidio Pharmaceuticals—underscores the complexities and high attrition rates in drug development. Despite initial promise, particularly against drug-resistant virus, the compound could not overcome its inherent risk-benefit limitations for chronic HIV therapy.
Nevertheless, the scientific understanding gained from Alovudine's development has had lasting implications. Its interaction with thymidine kinase and subsequent intracellular trapping formed the basis for its successful repurposing as 18F−FLT, a PET imaging agent used to measure cellular proliferation in oncology and other research areas. Furthermore, the very mechanism responsible for its dose-limiting toxicity in HIV (mitochondrial DNA polymerase gamma inhibition) has spurred investigation into its potential as an anti-leukemic agent, aiming to exploit this mitochondrial disruption as a therapeutic strategy against certain cancers.
In summary, Alovudine's story is a salient example of a drug candidate with clear biological activity that faced insurmountable challenges in achieving a safe and effective clinical profile for its primary indication. It highlights the critical importance of the therapeutic index, the complexities of off-target effects (particularly mitochondrial toxicity for NRTIs), and the dynamic nature of pharmaceutical development where scientific understanding can lead to alternative applications even for compounds that do not reach their initial therapeutic goals.
Works cited
- Alovudine | C10H13FN2O4 | CID 33039 - PubChem, accessed May 26, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Alovudine
- Buy Alovudine | 25526-93-6 | >98% - Smolecule, accessed May 26, 2025, https://www.smolecule.com/products/s518083
- Alovudine: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed May 26, 2025, https://go.drugbank.com/drugs/DB06198
- ALOVUDINE - Inxight Drugs, accessed May 26, 2025, https://drugs.ncats.io/drug/PG53R0DWDQ
- Alovudine - Wikipedia, accessed May 26, 2025, https://en.wikipedia.org/wiki/Alovudine
- ANTIVIRAL DRUGS IN THE TREATMENT OF AIDS: WHAT IS IN THE PIPELINE ?, accessed May 26, 2025, https://daignet.de/media/filer_public/a6/09/a609473b-49f1-4c83-84e3-079d6b34e420/stellbrink.pdf
- Mitochondrial DNA Depletion in Rat Liver Induced by Fosalvudine Tidoxil, a Novel Nucleoside Reverse Transcriptase Inhibitor Prodrug - PMC, accessed May 26, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2704664/
- The Kinetic Effects on Thymidine Kinase 2 by Enzyme-Bound dTTP May Explain the Mitochondrial Side Effects of Antiviral Thymidine Analogs | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed May 26, 2025, https://journals.asm.org/doi/10.1128/AAC.00109-11
- Mitochondrial DNA Depletion in Rat Liver Induced by Fosalvudine Tidoxil, a Novel Nucleoside Reverse Transcriptase Inhibitor Prodrug | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed May 26, 2025, https://journals.asm.org/doi/10.1128/aac.00364-09
- 25526-93-6 | Alovudine - ChemScene, accessed May 26, 2025, https://www.chemscene.com/25526-93-6.html
- Alovudine, accessed May 26, 2025, http://medbox.iiab.me/kiwix/wikipedia_en_medicine_2019-12/A/Alovudine
- The thymidine dideoxynucleoside analog, alovudine, inhibits the mitochondrial DNA polymerase γ, impairs oxidative phosphorylation and promotes monocytic differentiation in acute myeloid leukemia - PubMed Central, accessed May 26, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6518883/
- Alovudine - MedPath Clinical Trial Search, accessed May 26, 2025, https://trial.medpath.com/drug/86732d248b17b3db
- Medivir AB Outlicenses The Antiviral Compounds Alovudine (MIV-310) And MIV-410 To Presidio Pharmaceuticals - BioSpace, accessed May 26, 2025, https://www.biospace.com/medivir-ab-outlicenses-the-antiviral-compounds-alovudine-miv-310-and-miv-410-to-presidio-pharmaceuticals
- Biopharma collaborations in 2006 - | BioWorld, accessed May 26, 2025, https://www.bioworld.com/content/biotechbig-pharma-collaborations-2
- Alovudine | 25526-93-6 - Sigma-Aldrich, accessed May 26, 2025, https://www.sigmaaldrich.com/US/en/product/chemscenellcpreferredpartner/ciah98d1e0be?context=bbe
- Alovudine - MedPath Clinical Trial Search, accessed May 26, 2025, https://trial.medpath.com/drug/86732d248b17b3db/alovudine
- Alovudine | Antiviral - TargetMol, accessed May 26, 2025, https://www.targetmol.com/compound/3%27-fluoro-3%27-deoxythymidine
- MIV-310 reduces HIV viral load in patients failing multiple antiretroviral therapy: results from a 4-week phase II study - PubMed, accessed May 26, 2025, https://pubmed.ncbi.nlm.nih.gov/15362662/
- Antiviral activity of low-dose alovudine in antiretroviral-experienced patients: results from a 4-week randomized, double-blind, placebo-controlled dose-ranging trial - PubMed, accessed May 26, 2025, https://pubmed.ncbi.nlm.nih.gov/17461857/
- New Drug Targets for HIV, accessed May 26, 2025, https://academic.oup.com/cid/article-pdf/41/Supplement_1/S96/20907243/41-Supplement_1-S96.pdf
- ENDOCYTE, INC - Cancer Imaging Program, accessed May 26, 2025, https://imaging.cancer.gov/programs_resources/cancer-tracer-synthesis-resources/docs/flt_ib_msword.doc
- Antiviral activity of low-dose alovudine in antiretroviral-experienced patients: Results from a 4-week randomized, double-blind, placebo-controlled dose-ranging trial - ResearchGate, accessed May 26, 2025, https://www.researchgate.net/publication/6367120_Antiviral_activity_of_low-dose_alovudine_in_antiretroviral-experienced_patients_Results_from_a_4-week_randomized_double-blind_placebo-controlled_dose-ranging_trial
- Alovudine (3'-Fluoro-3'-deoxythymidine) | mtDNA Synthesis Inhibitor | MedChemExpress, accessed May 26, 2025, https://www.medchemexpress.com/alovudine.html
- Relationship between plasma concentrations of 3'-deoxy-3 ..., accessed May 26, 2025, https://pubmed.ncbi.nlm.nih.gov/7995977/
- An Ascending, Single Oral Dose Pharmacokinetic Study of 3'-Deoxy-3'-Fluorothymidine (FLT) in Asymptomatic Human Immunodeficiency Virus (HIV)-Infected Subjects - MedPath Clinical Trial Search, accessed May 26, 2025, https://trial.medpath.com/clinical-trial/84c0aba9f4e64734/nct00002271-pharmacokinetic-study-FLT-HIV-subjects
- Alovudine |CAS#25526-93-6 | reverse transcriptase inhibitor | HIV | MedKoo, accessed May 26, 2025, https://www.medkoo.com/products/12141
- Alovudine Medivir - PubMed, accessed May 26, 2025, https://pubmed.ncbi.nlm.nih.gov/12669386/
- accessed January 1, 1970, https://clinicaltrials.gov/study/NCT00002254
- (PDF) Mitochondrial DNA Depletion in Rat Liver Induced by Fosalvudine Tidoxil, a Novel Nucleoside Reverse Transcriptase Inhibitor Prodrug - ResearchGate, accessed May 26, 2025, https://www.researchgate.net/publication/24419075_Mitochondrial_DNA_Depletion_in_Rat_Liver_Induced_by_Fosalvudine_Tidoxil_a_Novel_Nucleoside_Reverse_Transcriptase_Inhibitor_Prodrug
- Toxicology evaluation of radiotracer doses of 3'-deoxy-3'-[18F]fluorothymidine (18F-FLT) for human PET imaging: Laboratory analysis of serial blood samples and comparison to previously investigated therapeutic FLT doses - PMC - PubMed Central, accessed May 26, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC1931583/
- Alovudine F-18 Terminated Phase 1 Trials for Central Nervous System Neoplasm Diagnostic | DrugBank Online, accessed May 26, 2025, https://go.drugbank.com/drugs/DB14930/clinical_trials?conditions=DBCOND0055396&phase=1&purpose=diagnostic&status=terminated
- PET/CT with 18F-FLT Is Unlikely to Cause Significant Hepatorenal or Hematologic Toxicity - Journal of Nuclear Medicine, accessed May 26, 2025, https://jnm.snmjournals.org/content/jnumed/51/5/824.2.full.pdf