C45H54F2N4O8
162652-95-1
Transitional Cell Carcinoma of the Urothelial Tract, Metastatic Transitional Cell Carcinoma of the Urothelial Tract
Vinflunine is a third-generation, fluorinated vinca alkaloid that represents an incremental but important evolution in its therapeutic class. Its primary clinical value is established by its European Medicines Agency (EMA) approval as a second-line monotherapy for advanced or metastatic urothelial carcinoma after the failure of platinum-based chemotherapy, a clinical niche now significantly challenged by the advent of newer immunotherapies and antibody-drug conjugates (ADCs). The unique pharmacological profile of vinflunine, characterized by a distinct and weaker interaction with tubulin, may confer a more favorable neurotoxicity profile compared to its predecessors. This potential safety advantage is counterbalanced by a modest survival benefit and a significant, though manageable, safety burden dominated by myelosuppression and constipation. The divergent regulatory decisions by the EMA, which granted approval, and the United States Food and Drug Administration (FDA), which did not, underscore the fine balance of its risk-benefit profile in a rapidly evolving oncology landscape.
Key characteristics of vinflunine include:
Vinflunine is a small molecule drug classified as a third-generation vinca alkaloid.[1] It is marketed under the brand name Javlor®.[1] Its International Nonproprietary Name (INN) is vinflunine, with international variations including
vinflunina and vinfluninum.[1] Chemically, it is identified by several synonyms, most notably 20',20'-difluoro-3',4'-dihydrovinorelbine, 4'-deoxy-20',20'-difluoro-5'-norvincaleukoblastine, and 4'-deoxy-20',20'-difluoro-8'-norvincaleukoblastine.[1]
For precise identification in scientific and regulatory databases, vinflunine is assigned multiple unique identifiers. Its Chemical Abstracts Service (CAS) Registry Number is 162652-95-1.[1] Other key identifiers include its DrugBank Accession Number (DB11641), FDA Unique Ingredient Identifier (UNII) (5BF646324K), ChEBI ID (CHEBI:90241), and NCI Thesaurus Code (C61564).[1] In the Anatomical Therapeutic Chemical (ATC) classification system, it is coded as L01CA05, placing it within the Vinca alkaloids and analogues subclass of antineoplastic agents.[8]
Vinflunine is a complex organic heteropentacyclic and heterotetracyclic compound with the molecular formula C45H54F2N4O8 and a molecular weight of approximately 816.94 g/mol.[11] It is a semi-synthetic derivative of vinorelbine, another vinca alkaloid, distinguished by the strategic introduction of two fluorine atoms.[8] This structural modification is achieved through a novel synthesis process using superacid media (
HF−SbF5) on anhydrovinblastine, which allows for the selective difluorination of the catharanthine moiety of the molecule—a region previously inaccessible to chemical modification.[12] This bi-fluorination is central to its unique pharmacological properties.
The drug is supplied for clinical use as vinflunine ditartrate, a salt form that enhances its stability and solubility.[8] Physically, it is a solid that is formulated as a clear, colorless to pale yellow sterile concentrate (25 mg/mL) intended for intravenous infusion after dilution.[17]
Table 1: Key Drug Identifiers and Chemical Properties | |
---|---|
Parameter | Value |
Generic Name | Vinflunine |
Brand Name | Javlor® |
DrugBank ID | DB11641 |
CAS Number | 162652-95-1 |
Molecular Formula | C45H54F2N4O8 |
Molecular Weight | 816.94 g/mol |
Chemical Class | Third-generation Vinca Alkaloid |
IUPAC Name | methyl (2β,3β,4β,5α,12β,19α)- 4-(acetyloxy)- 15-indol- 8-yl]- 3-hydroxy- 16-methoxy- 1-methyl- 6,7-didehydroaspidospermidine- 3-carboxylate |
Vinflunine exerts its antineoplastic effects as a potent anti-mitotic agent, a hallmark of the vinca alkaloid class.[1] Its primary molecular target is tubulin, the protein heterodimer that polymerizes to form microtubules.[16] Microtubules are critical components of the cellular cytoskeleton, responsible for maintaining cell structure, facilitating intracellular transport, and, most importantly, forming the mitotic spindle required for chromosome segregation during cell division.[1]
Vinflunine binds at or near the established vinca-binding sites on the β-tubulin subunit, likely at the interface between tubulin dimers.[1] This binding event inhibits the polymerization of tubulin into functional microtubules.[1] More specifically, vinflunine disrupts the exquisitely regulated and highly dynamic nature of the microtubule network. It suppresses two key processes:
By disrupting these dynamics, vinflunine destabilizes the mitotic spindle apparatus. This prevents the proper alignment and segregation of chromosomes, leading to a cell cycle arrest at the G2/M phase, specifically at the metaphase/anaphase transition.[1] The prolonged mitotic arrest activates cellular checkpoints that ultimately trigger apoptosis, or programmed cell death, thereby eliminating the proliferating cancer cells.[1] In addition to its direct anti-mitotic effects, vinflunine has also been shown to possess antiangiogenic and antivascular properties, which may contribute to its overall antitumor activity by disrupting the formation of new blood vessels that supply tumors.[14]
The pharmacodynamic profile of vinflunine is distinguished from that of its predecessors, vinblastine and vincristine. A central paradox of its pharmacology is that while it demonstrates superior antitumor activity in preclinical in vivo models, its binding affinity for the vinca-binding site on tubulin is significantly weaker.[1] This seemingly contradictory observation is fundamental to understanding its clinical profile. The weaker, more readily reversible binding to tubulin is hypothesized to be the structural basis for its potentially improved safety profile, particularly with respect to neurotoxicity.[8] Older vinca alkaloids are notorious for causing dose-limiting peripheral neuropathy due to their disruption of the stable axonal microtubules in neurons. The less tenacious binding of vinflunine may be sufficient to disrupt the highly sensitive and dynamic mitotic spindle in rapidly dividing cancer cells but less damaging to the more stable microtubule structures in non-dividing neuronal cells. This differential effect may create a wider therapeutic window, allowing for a dose and exposure level that is highly cytotoxic to tumors while being better tolerated by the peripheral nervous system. This optimized balance between efficacy and toxicity, rather than raw binding potency, likely explains its superior
in vivo performance in preclinical models.[1]
This unique interaction with tubulin may also explain why vinflunine is a less-potent inductor of P-glycoprotein-mediated multidrug resistance in vitro compared to vinorelbine.[1] The reduced selective pressure from a weaker-binding agent may slow the emergence of resistance mechanisms. In patients, the pharmacodynamic effects of vinflunine are primarily observed as concentration- and exposure-dependent toxicities, with myelosuppression (particularly neutropenia) and gastrointestinal effects (constipation) being the most prominent.[1]
As vinflunine is administered exclusively via the intravenous route, the absorption phase of its pharmacokinetics is bypassed, ensuring 100% bioavailability. The drug exhibits a linear pharmacokinetic profile across a wide range of clinically relevant doses, from 30 mg/m² to 400 mg/m², meaning that exposure increases proportionally with the dose.[1]
Distribution: Vinflunine is characterized by extensive distribution into the tissues. This is evidenced by its very large terminal volume of distribution, which has been measured at 2422 ± 676 L, corresponding to approximately 35 L/kg.[1] This extensive tissue penetration is a key factor in its pharmacological activity. The drug is moderately bound to human plasma proteins, with a binding fraction of 67.2% to 68.3%.[1] It primarily binds to high-density lipoproteins (HDL) and serum albumin, with negligible binding to alpha-1 acid glycoprotein or platelets.[1] This binding is non-saturable within the range of concentrations observed in patients.[1] The ratio of vinflunine concentration in plasma versus whole blood is approximately 0.80.[1]
Metabolism: Vinflunine undergoes significant metabolism in the body. The primary enzyme responsible for its Phase I metabolism is the cytochrome P450 isoenzyme CYP3A4.[12] In addition to CYP3A4-mediated pathways, vinflunine is also metabolized by various esterases. This esterase activity leads to the formation of 4-O-deacetylvinflunine (DVFL), which is the only known active metabolite of vinflunine and is the main metabolite detected in the bloodstream.[12] The prolonged presence of this active metabolite contributes significantly to the overall therapeutic and toxic effects of the drug.
Excretion: The elimination of vinflunine and its metabolites occurs through two main routes. The majority of the drug and its byproducts are eliminated via the hepatobiliary system, with fecal excretion accounting for approximately two-thirds of the total clearance.[1] The remaining one-third is excreted renally via the urine.[1] The clearance kinetics are characterized by a mean terminal half-life of approximately 40 hours for the parent vinflunine molecule.[1] Notably, the active metabolite DVFL has a significantly longer half-life of approximately 120 hours, leading to sustained exposure and pharmacological activity well after the parent drug concentrations have declined.[1]
Table 2: Summary of Pharmacokinetic Parameters for Vinflunine and its Active Metabolite (DVFL) | |
---|---|
Parameter | Value |
Volume of Distribution (Vd) | ~2422 L (~35 L/kg) |
Plasma Protein Binding | 67.2–68.3% |
Primary Metabolism Route | CYP3A4 and Esterases |
Active Metabolite | 4-O-deacetylvinflunine (DVFL) |
Terminal Half-life (Vinflunine) | ~40 hours |
Terminal Half-life (DVFL) | ~120 hours |
Primary Excretion Route | Fecal (~67%) and Renal (~33%) |
The regulatory approval of vinflunine in Europe was predicated on a clinical development program that culminated in a pivotal, multinational, randomized Phase III trial.[4] This study was designed to evaluate the efficacy and safety of vinflunine plus Best Supportive Care (BSC) compared to BSC alone in patients with advanced or metastatic transitional cell carcinoma of the urothelial tract (TCCU) whose disease had progressed following a first-line platinum-containing chemotherapy regimen.[4] This was a critical unmet need, as no standard second-line therapy existed at the time.
The trial enrolled 370 patients who were randomized in a 2:1 ratio to either the vinflunine plus BSC arm or the BSC alone arm.[4] The primary endpoint of the study was overall survival (OS).[4] The dosing regimen for vinflunine was initially set at 320 mg/m² every three weeks. However, following an early safety review, the protocol was amended to a risk-stratified approach: patients with a good performance status (ECOG PS=0) and no prior pelvic irradiation received the 320 mg/m² dose, while patients with a performance status of 1 or with prior pelvic radiation started at a reduced dose of 280 mg/m², with the option to escalate to 320 mg/m² if the first cycle was well tolerated.[4] This trial was preceded by two Phase II studies that established the initial signal of activity for vinflunine in this patient population, providing the rationale for the definitive Phase III investigation.[7]
The results of the pivotal Phase III trial presented a nuanced efficacy profile. In the primary analysis of the intent-to-treat (ITT) population (all 370 randomized patients), the median overall survival was 6.9 months in the vinflunine arm compared to 4.6 months in the BSC arm.[4] While this represented a 2.3-month improvement, the difference did not achieve statistical significance (hazard ratio for death = 0.88; 95% CI, 0.69 to 1.12;
p=0.287).[4]
However, the trial's statistical plan included pre-specified analyses of the "eligible" patient population (n=357), which excluded patients who did not meet all eligibility criteria. In this population, the median OS was 6.9 months for vinflunine versus 4.3 months for BSC, a difference that was statistically significant (HR = 0.78; 95% CI, 0.61 to 0.96; p=0.040).[4] Furthermore, a multivariate Cox regression analysis, which adjusted for baseline prognostic factors that were imbalanced between the arms (such as performance status), demonstrated a statistically significant treatment effect for vinflunine, corresponding to a 23% reduction in the risk of death (HR = 0.77; 95% CI, 0.61 to 0.98;
p=0.036).[4]
Secondary endpoints consistently favored the vinflunine arm. The overall response rate (ORR), disease control rate (DCR), and progression-free survival (PFS) were all statistically significantly improved in patients receiving vinflunine compared to those receiving BSC alone (p=0.006, p=0.002, and p=0.001, respectively).[4] Subsequent long-term follow-up of the trial confirmed these findings, reinforcing the survival benefit observed in the eligible population.[5] Real-world data from post-marketing studies have generally corroborated these results, with retrospective analyses from Spanish centers reporting an ORR of 25.5% and a median OS of 10 months in a cohort of 102 patients.[27]
Table 3: Efficacy Outcomes from the Pivotal Phase III Trial (Bellmunt et al., 2009) | Vinflunine + BSC Arm | BSC Alone Arm | Statistics |
---|---|---|---|
Intent-to-Treat (ITT) Population (n=370) | |||
Median Overall Survival (OS) | 6.9 months | 4.6 months | HR = 0.88; p=0.287 |
Eligible Population (n=357) | |||
Median Overall Survival (OS) | 6.9 months | 4.3 months | HR = 0.78; p=0.040 |
Secondary Endpoints (ITT Population) | |||
Overall Response Rate (ORR) | 8.6% | 0% | p=0.006 |
Disease Control Rate (DCR) | 41.1% | 24.8% | p=0.002 |
Median Progression-Free Survival (PFS) | 3.0 months | 1.5 months | p=0.001 |
The clinical history of vinflunine serves as a compelling case study in the rapid evolution of cancer therapeutics. Upon its approval in 2009, it became a landmark agent, establishing the first evidence-based standard of care for second-line TCCU in Europe.[7] However, its position has been successively challenged and largely superseded by newer, more effective classes of drugs.
Comparison with Taxanes: In the second-line setting, taxanes such as docetaxel and paclitaxel have been used off-label. A direct comparison was undertaken in the SECAVIN trial, which randomized patients to receive either vinflunine or the taxane cabazitaxel.[29] The study was terminated after the Phase II stage due to a lack of efficacy in the cabazitaxel arm. The results showed a trend favoring vinflunine, with a significantly longer median PFS (2.9 vs. 1.9 months;
p=0.039) and a numerically longer median OS (7.6 vs. 5.5 months).[29] This suggested that within the realm of cytotoxic chemotherapy, vinflunine held an advantage over at least one potent taxane in this specific indication.
Displacement by Immune Checkpoint Inhibitors (ICIs): The therapeutic landscape was transformed by the introduction of immune checkpoint inhibitors. The pivotal KEYNOTE-045 trial directly compared the anti-PD-1 antibody pembrolizumab against the investigator's choice of chemotherapy (paclitaxel, docetaxel, or vinflunine) in the same second-line, platinum-refractory setting.[30] The trial demonstrated a clear and significant overall survival benefit for pembrolizumab, which has since become the preferred standard of care in this setting where available, effectively displacing vinflunine and other chemotherapies from their primary role.[31]
Displacement by Antibody-Drug Conjugates (ADCs): For patients who progress on both platinum-based chemotherapy and an ICI, the Nectin-4-directed ADC enfortumab vedotin has emerged as a new standard of care. The EV-301 trial randomized such patients to receive either enfortumab vedotin or chemotherapy (including vinflunine).[33] The results were definitive, showing a significant survival advantage for enfortumab vedotin, with a median OS of 12.88 months compared to 8.97 months for chemotherapy (HR = 0.70;
p=0.001).[33]
Current Role and Post-ICI Activity: As a result of these developments, vinflunine's role has been relegated to later lines of therapy or in regions where newer agents are not accessible. However, recent retrospective data suggest it retains meaningful clinical activity even in heavily pre-treated patients. A German multicenter analysis of patients treated with vinflunine found that those who had previously received an ICI (post-ICI cohort) had a significantly higher clinical benefit rate (CBR) compared to ICI-naïve patients (51.0% vs. 25.0%; p=0.020) and showed a trend towards improved OS (8.78 vs. 5.72 months).[35] This suggests that vinflunine remains a viable therapeutic option in the third-line or later setting for select patients, and there is no evidence of cross-resistance with immunotherapy.
The administration of vinflunine requires careful adherence to established protocols to maximize efficacy and minimize toxicity. The standard recommended dose for eligible adult patients is 320 mg/m², calculated based on the patient's body surface area.[37] This dose is administered as a 20-minute intravenous infusion, repeated once every three weeks.[37]
It is imperative that vinflunine is administered only via the intravenous route. Inadvertent intrathecal administration is potentially fatal and is strictly contraindicated.[37] The drug is supplied as a concentrate that must be diluted prior to administration. The calculated volume of concentrate is typically added to a 100 ml infusion bag containing either 0.9% sodium chloride solution or 5% glucose solution.[37] To mitigate the risk of venous irritation and extravasation, a specific administration procedure is recommended. This involves establishing venous access (preferably via a central line or a large vein in the upper forearm), flushing the vein with at least an equal volume of infusion solution both before and after the 20-minute vinflunine infusion.[18]
Vinflunine has a narrow therapeutic index, necessitating proactive dose adjustments for specific patient populations and in response to treatment-related toxicities.[38] The approved labeling provides detailed guidance for dose modifications.
Table 4: Recommended Dose Adjustments for Vinflunine | |
---|---|
Condition | Recommended Dose (mg/m²) |
Standard Dose (PS=0, no prior pelvic radiation) | 320 |
ECOG PS=1 / Prior Pelvic Radiation | 280 (initial dose) |
Elderly (75-79 years) | 280 |
Elderly (≥80 years) | 250 |
Moderate Renal Impairment (CrCl 40-60 ml/min) | 280 |
Severe Renal Impairment (CrCl 20-40 ml/min) | 250 |
Mild Hepatic Impairment (Child-Pugh A) | 250 |
Moderate Hepatic Impairment (Child-Pugh B) | 200 |
Grade 4 Neutropenia >7 days (First Event) | Reduce to 280 (if initial dose was 320) |
Proactive patient management is essential to mitigate the predictable toxicities of vinflunine. Due to the high incidence and potential severity of constipation, prophylactic measures are strongly recommended for all patients.[37] This includes dietary counseling (e.g., adequate oral hydration, increased fiber intake) and the routine prescription of laxatives (e.g., osmotic or stimulant laxatives) to be taken from day 1 through day 5 or 7 of each treatment cycle.[37]
Given the high rates of hematological toxicity, complete blood counts, including absolute neutrophil count (ANC), platelet count, and hemoglobin levels, must be monitored before every infusion of vinflunine.[37] Treatment should be delayed if hematological parameters do not meet the specified safety thresholds.[37]
The safety profile of vinflunine is well-characterized and consistent with its mechanism as a cytotoxic microtubule inhibitor. The most frequently reported adverse events are hematological and gastrointestinal in nature.[39]
Table 5: Profile of Common (≥10%) and Serious Adverse Reactions (Grade ≥3) | ||
---|---|---|
Adverse Reaction (by System Organ Class) | Incidence (All Grades %) | Incidence (Grade 3-4 %) |
Blood and Lymphatic System Disorders | ||
Anaemia | 92.8% | 17.3-19% |
Leucopenia | 84.5% | 45.2% |
Neutropenia | 79.6% | 50-54.6% |
Thrombocytopenia | 53.5% | 4.9% |
Febrile Neutropenia | 6.7% | 6-6.7% |
Gastrointestinal Disorders | ||
Constipation | 54.9% | 15.3-16% |
Nausea | 40.9% | 2.9% |
Stomatitis / Mucositis | 26.9% | 2.7% |
Abdominal Pain | 21.6% | 4.7% |
General Disorders | ||
Asthenia / Fatigue | 55.3% | 15.8-19% |
Nervous System Disorders | ||
Peripheral Sensory Neuropathy | 11.3% | 0.9% |
The use of vinflunine is subject to several absolute contraindications and important warnings to ensure patient safety.
As vinflunine is a substrate of the CYP3A4 enzyme, its pharmacokinetics can be significantly altered by co-administration of drugs that inhibit or induce this pathway.[12]
Vinflunine was discovered at the Pierre Fabre research center in France in collaboration with the team of Professor Jean-Claude Jacquesy at the University of Poitiers.[1] First described in 1998, its development was a result of a targeted effort to improve upon the existing vinca alkaloids, particularly its parent compound, vinorelbine.[1] The key chemical innovation was the application of superacidic chemistry, which enabled the selective introduction of two fluorine atoms at the 20' position of the catharanthine moiety of the molecule.[12] This structural modification was crucial, as it altered the drug's interaction with tubulin and was hypothesized to be the basis for an improved therapeutic profile.[15]
Preclinical development confirmed this hypothesis. In vitro and in vivo studies demonstrated that vinflunine possessed marked antitumor activity across a broad range of experimental models, often superior to that of vinorelbine.[1] Importantly, these studies also suggested a more favorable safety profile, with evidence of reduced neurotoxicity and a lower propensity to induce drug resistance mechanisms compared to older vinca alkaloids.[23] These promising preclinical data provided a strong rationale for advancing vinflunine into clinical trials for human cancers.[45]
The clinical development of vinflunine was initially a collaborative effort. In 2004, Pierre Fabre licensed the development and commercialization rights for certain territories, including the United States, to Bristol-Myers Squibb (BMS).[47] However, this partnership was terminated in November 2007, with BMS halting its development plans for the drug in the US.[43] Pierre Fabre reacquired the global rights and continued to pursue regulatory approval independently, focusing on the European market.[43]
This strategy proved successful. On September 21, 2009, the European Medicines Agency (EMA) granted marketing authorization for vinflunine, under the trade name Javlor, for its indication in second-line advanced or metastatic TCCU.[7] The drug is also registered in other jurisdictions, such as Australia.[43]
A pivotal aspect of vinflunine's history is its divergent regulatory outcomes in major markets. Despite its approval in Europe, vinflunine did not receive approval from the US Food and Drug Administration (FDA).[7] This split decision likely stems from different interpretations of the pivotal Phase III trial data. The trial's primary endpoint—overall survival in the intent-to-treat (ITT) population—failed to meet the threshold for statistical significance (
p=0.287).[4] For a regulatory body like the FDA, which often places stringent emphasis on the pre-specified primary endpoint in the ITT population, this result would present a significant hurdle for approval. In contrast, the EMA's decision appears to have been based on a "totality of the evidence" approach. The agency likely gave more weight to the statistically significant survival benefit observed in the pre-specified "eligible" patient population (
p=0.040), the positive results from the multivariate analysis that corrected for baseline imbalances, and the consistent, statistically significant improvements across all secondary efficacy endpoints.[4] This case highlights how differences in regulatory philosophy and the interpretation of complex clinical trial data can lead to a drug becoming a standard of care in one region while remaining unavailable in another.
Vinflunine represents a product of rational drug design, a carefully engineered modification of a classic chemotherapeutic scaffold that successfully translated into a distinct pharmacological profile. At the time of its approval, it filled a critical void in the treatment of advanced urothelial carcinoma, offering the first evidence-based survival benefit for patients who had progressed on platinum-based chemotherapy. Its story is one of incremental innovation, providing a valuable, albeit temporary, standard of care.
The modest magnitude of its survival benefit, coupled with a significant but predictable and manageable toxicity profile, defined its clinical utility. However, the rapid pace of innovation in oncology, particularly the advent of highly effective immune checkpoint inhibitors and antibody-drug conjugates, has fundamentally reshaped the treatment paradigm for urothelial cancer. These newer agents have demonstrated superior efficacy, leading to the displacement of vinflunine from its primary second-line indication. Nonetheless, its retained clinical activity in heavily pre-treated patients, including those who have progressed on immunotherapy, suggests that vinflunine may still hold a niche role as a salvage therapy in later lines of treatment, particularly in healthcare systems where access to newer, more expensive agents is limited.
While its primary indication has been challenged, research into vinflunine continues, exploring its potential in other clinical contexts. Recently completed or ongoing clinical trials have investigated its use in other malignancies, such as in combination with capecitabine for advanced breast cancer (NCT01095003) and as a monotherapy for advanced carcinoma of the penis (VinCaP trial, NCT02057913).[49] Studies have also explored novel combinations, such as with the multi-kinase inhibitor sorafenib (NCT01844947) or with gemcitabine for cisplatin-ineligible patients (VINGEM trial, NCT02665039), in an effort to enhance its efficacy or expand its use to the first-line setting for specific subgroups.[51]
The most critical question for the future of vinflunine lies in defining its optimal place in the modern, complex sequencing of therapies for urothelial carcinoma. As more patients are exposed to both chemotherapy, ICIs, and ADCs, understanding the efficacy and safety of vinflunine in the third- or fourth-line setting will be crucial. The preliminary evidence suggesting a potential for enhanced benefit in post-ICI patients warrants further prospective investigation.[35] The future of vinflunine is not as a frontline agent but as a potential salvage therapy option for a carefully selected, heavily pre-treated patient population, where its unique mechanism and manageable safety profile may still offer meaningful clinical benefit.
Published at: July 31, 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.