Small Molecule
C17H14O4
117570-53-3
Brivanib alaninate (BMS-582664) is an orally bioavailable, investigational small molecule designed as a targeted therapy for solid tumors. It is the L-alanine ester prodrug of its active moiety, brivanib (BMS-540215), a potent, ATP-competitive dual inhibitor of Vascular Endothelial Growth Factor Receptor 2 (VEGFR-2) and Fibroblast Growth Factor Receptor 1 (FGFR-1).[1] The development of brivanib alaninate was predicated on a strong scientific rationale, particularly for the treatment of advanced Hepatocellular Carcinoma (HCC), a malignancy where both the VEGF and FGF signaling pathways are known to be critical drivers of tumor angiogenesis, proliferation, and survival. The central hypothesis was that simultaneous inhibition of both pathways could provide a more robust and durable antitumor effect and potentially overcome the adaptive resistance mechanisms that limit the efficacy of therapies targeting only the VEGF pathway.[4]
Developed by Bristol-Myers Squibb, brivanib alaninate underwent an extensive global clinical development program that enrolled over 4,000 patients across a range of malignancies, including colorectal, cervical, and endometrial cancers, with a primary strategic focus on advanced HCC.[7] Despite promising preclinical data and encouraging results from Phase II studies, the compound's trajectory was ultimately defined by the negative outcomes of two pivotal, large-scale Phase III trials in HCC.
The BRISK-FL study (NCT00858871), which evaluated brivanib alaninate against the standard-of-care sorafenib in the first-line treatment of advanced HCC, failed to meet its primary endpoint of demonstrating non-inferiority in Overall Survival (OS).[9] Concurrently, the BRISK-PS study (NCT00825955), assessing the drug against placebo in patients who had progressed on or were intolerant to sorafenib, also failed to show a statistically significant improvement in OS, despite achieving significant benefits in secondary endpoints such as Time to Progression (TTP) and Objective Response Rate (ORR).[11]
The clinical safety profile of brivanib alaninate was characterized as manageable but was ultimately less well-tolerated than sorafenib. It was associated with higher rates of clinically significant adverse events, including fatigue, hypertension, and hyponatremia, which led to a notably higher rate of treatment discontinuation compared to the standard of care.[9] This unfavorable therapeutic index is considered a primary contributor to its clinical failure. Consequently, following the definitive results of the Phase III program, the orphan drug designations granted by U.S. and European regulatory agencies were withdrawn, and Bristol-Myers Squibb discontinued the development of brivanib alaninate.[1] The history of brivanib alaninate serves as an important case study in oncology drug development, illustrating the significant challenges in translating a compelling biological hypothesis into a clinically meaningful survival benefit, and underscoring the critical importance of the therapeutic window in determining the ultimate success or failure of a novel agent.
Brivanib alaninate is a synthetic organic compound classified as a small molecule drug.[16] Structurally, it is a complex heterocyclic molecule belonging to the pyrrolotriazine chemical class and is further characterized as a fluoroindole, an aromatic ether, a diether, and a carboxylic ester, specifically an L-alanine derivative.[1] Its formal chemical name, established by the International Union of Pure and Applied Chemistry (IUPAC), is-5-methylpyrrolo[2,1-f]triazin-6-yl]oxypropan-2-yl] (2S)-2-aminopropanoate.[1] During its development by Bristol-Myers Squibb, it was primarily identified by the code BMS-582664.[2]
The compound's fundamental properties have been well-characterized. It possesses a molecular formula of and an average molecular mass of 441.463 g/mol.[16] Its physicochemical characteristics are critical to its behavior as a drug. It has very low aqueous solubility, measured at 0.0127 mg/mL, a property that was a central challenge in its development.[16] Calculated lipophilicity (logP) values range from 2.45 to 3.5, indicating a moderately lipophilic nature.[16] From a drug-likeness perspective, brivanib alaninate exhibits favorable properties, conforming to Lipinski's Rule of Five and the Ghose Filter, which are computational filters used to predict if a chemical compound has properties that would make it a likely orally active drug in humans.[16]
Table 1: Key Chemical and Physical Properties of Brivanib Alaninate
| Property | Value | Source(s) |
|---|---|---|
| DrugBank ID | DB11865 | 1 |
| Type | Small Molecule | 16 |
| CAS Number | 649735-63-7 | 1 |
| IUPAC Name | -5-methylpyrrolo[2,1-f]triazin-6-yl]oxypropan-2-yl] (2S)-2-aminopropanoate | 1 |
| InChIKey | LTEJRLHKIYCEOX-OCCSQVGLSA-N | 1 |
| SMILES | CC1=CC2=C(N1)C=CC(=C2F)OC3=NC=NN4C3=C(C(=C4)OCC@@HOC(=O)N)C | |
| Molecular Formula | ||
| Average Mass | 441.463 g/mol | |
| Water Solubility | 0.0127 mg/mL | |
| logP | 2.45 (ALOGPS), 3.5 (Chemaxon) | |
| Hydrogen Bond Acceptors | 5 | |
| Hydrogen Bond Donors | 2 | |
| Rotatable Bonds | 8 | |
| Polar Surface Area | 116.76 | |
| Lipinski's Rule of Five | Yes (0 violations) |
The chemical identity of brivanib alaninate is intrinsically linked to its strategic design as a prodrug. A prodrug is a pharmacologically inactive compound that is converted into an active drug within the body through metabolic processes. Brivanib alaninate is the L-alanine ester prodrug of the active pharmacological moiety, brivanib (BMS-540215), a secondary alcohol that is the potent dual VEGFR-2/FGFR-1 kinase inhibitor.
This medicinal chemistry strategy was not arbitrary but was a deliberate and necessary solution to a significant drug development challenge: the poor physicochemical properties of the parent compound, brivanib. Preclinical studies revealed that brivanib suffered from very low aqueous solubility, which in turn led to dissolution rate-limited absorption and variable, often poor, oral bioavailability. To create a viable oral therapeutic, chemists prepared a series of amino acid ester prodrugs with the goal of improving these properties. The formal condensation of the carboxy group of the amino acid L-alanine with the hydroxy group of brivanib created the ester linkage of brivanib alaninate. This modification dramatically improved the compound's properties, allowing for the use of completely aqueous vehicles for formulation and, most critically, leading to a substantial and consistent improvement in oral bioavailability. In preclinical species, this strategy increased bioavailability from a range of 22-88% for the parent drug to a much more favorable 55-97% for the prodrug form. This successful optimization of drug delivery was the key chemical innovation that enabled the extensive clinical investigation of the molecule.
The transition from a laboratory-scale chemical concept to a commercial-scale manufacturing process for brivanib alaninate introduced its own set of complex challenges. The very ester linkage that was crucial for improving bioavailability also rendered the active pharmaceutical ingredient (API) susceptible to hydrolysis, requiring a robust and tightly controlled manufacturing strategy to ensure product stability and purity.
The commercial synthesis process required meticulous control over both input materials and process-related impurities. Three aspects of the manufacturing control strategy were particularly significant. First, a deep understanding of reaction parameters was necessary to minimize the formation of a key regioisomeric impurity during the alkylation step with (R)-propylene oxide. Second, statistical models and a design space approach were developed to precisely control the formation of multiple impurities throughout the synthesis. Third, advanced process analytical technologies (PAT), such as in-situ Fourier-transform infrared spectroscopy (FT-IR), were implemented for real-time monitoring of critical reaction steps, such as the deprotection of the carbobenzyloxy (Cbz) group, to ensure the reaction proceeded to completion without degrading the sensitive product. These sophisticated manufacturing controls highlight the inherent tension in the drug's design: the chemical modification that solved the bioavailability problem simultaneously introduced a new set of challenges related to chemical stability and manufacturing complexity, which had to be overcome to produce a consistent, high-quality drug product for clinical use.
The pharmacological activity of brivanib alaninate resides entirely in its active moiety, brivanib, which functions as a potent and selective inhibitor of specific receptor protein-tyrosine kinases (EC 2.7.10.1). Brivanib is an ATP-competitive inhibitor, meaning it binds to the ATP-binding pocket of the kinase domain, thereby preventing the phosphorylation of downstream substrates and blocking signal transduction.
Its primary characteristic is its dual inhibitory profile. It demonstrates potent, high-affinity inhibition of Vascular Endothelial Growth Factor Receptor 2 (VEGFR-2), with a half-maximal inhibitory concentration () of 25 nM. VEGFR-2 is the principal mediator of the pro-angiogenic signals of VEGF and is almost exclusively expressed on vascular endothelial cells, making it a highly validated target for antiangiogenic cancer therapy. In addition to its potent activity against VEGFR-2, brivanib also exhibits moderate potency against Fibroblast Growth Factor Receptor 1 (FGFR-1), with an of 148 nM, as well as VEGFR-1 ( = 380 nM) and VEGFR-3 ( = 10 nM). The drug is highly selective; its inhibitory activity against other related tyrosine kinases, such as Platelet-Derived Growth Factor Receptor β (PDGFRβ) and Epidermal Growth Factor Receptor (EGFR), is more than 240-fold weaker, indicating a focused mechanism of action. This dual-targeting profile classifies brivanib as a multi-targeted antiangiogenesis agent, a VEGFR inhibitor, and an FGFR antagonist.
The inhibition of VEGFR and FGFR by brivanib translates into distinct and synergistic antitumor effects at the cellular and tissue levels.
The strategic decision to develop a dual VEGFR/FGFR inhibitor was firmly grounded in the molecular biology of HCC and the emerging understanding of resistance mechanisms to targeted therapies. Both the VEGF and FGF signaling pathways are well-established as being pathologically hyperactivated in HCC, where they collaboratively drive tumor growth, progression, and neovascularization.
A critical scientific hypothesis that gained prominence during the development of brivanib was the concept of adaptive or evasive resistance to antiangiogenic therapies. It was observed that tumors treated with agents that selectively block only the VEGF pathway could escape inhibition by up-regulating alternative proangiogenic signaling pathways. The FGF/FGFR axis was identified as one of the most prominent of these escape mechanisms. Tumors under the selective pressure of VEGF blockade could switch their dependence to FGF signaling to maintain their blood supply and continue to grow. Therefore, the development of brivanib was based on the compelling rationale that simultaneously blocking both VEGFR and FGFR would provide a more comprehensive and durable antiangiogenic blockade. This dual-targeting approach was hypothesized to not only inhibit tumor growth from the outset but also to pre-emptively block a key escape pathway, thereby preventing or overcoming resistance and leading to superior clinical outcomes compared to a VEGF-selective inhibitor like sorafenib.
This strong scientific rationale was substantiated by robust preclinical data. In multiple in vivo models using patient-derived HCC tumor xenografts implanted in mice, orally administered brivanib demonstrated broad-spectrum and significant antitumor activity, effectively suppressing tumor growth compared to control groups. The observed growth inhibition was directly linked to the drug's proposed mechanism of action, with analyses of treated tumors revealing increased rates of apoptosis, reduced microvessel density, and decreased cell proliferation.
Crucially, these preclinical studies provided direct evidence supporting the dual-targeting hypothesis. The sensitivity of the HCC xenograft lines to brivanib-induced growth inhibition was found to be positively correlated with the tumors' expression levels of FGFR-1 and FGFR-2. This finding strongly suggested that the drug's activity against the FGFR target was a meaningful contributor to its overall efficacy, not merely an ancillary pharmacological property. This body of evidence provided a solid and compelling preclinical proof-of-concept, forming a strong rationale for advancing brivanib alaninate into large-scale clinical trials for patients with HCC. The subsequent failure of the drug in Phase III, despite this robust preclinical foundation, highlights the profound challenges of translating even the most well-supported biological hypotheses into tangible clinical benefits, often due to an inability to achieve an adequate therapeutic window where efficacy can be realized without dose-limiting toxicities in human patients.
The clinical pharmacology of brivanib alaninate is defined by its function as a prodrug, with its pharmacokinetic (PK) profile being characterized by the rapid and efficient in vivo generation of its active moiety, brivanib.
Table 2: Summary of Brivanib (Active Moiety) Pharmacokinetic Parameters in Humans (Single 800 mg Oral Dose)
| Parameter | Value | Source(s) |
|---|---|---|
| (Median) | 1 hour | |
| (Geometric Mean) | 6146 ng/mL | |
| Terminal Half-life (Mean) | 13.8 - 14 hours | |
| Apparent Oral Clearance (Geometric Mean) | 14.7 L/h | |
| Primary Route of Elimination | Fecal | |
| Fecal Recovery (%) | 81.5% - 82% | |
| Urinary Recovery (%) | 12.2% |
Population pharmacokinetic analyses, which use data from a broad range of patients to model drug behavior and identify sources of variability, were conducted using data from 167 subjects in Phase I and II studies. These analyses confirmed that the PK of brivanib could be well-described by a linear, two-compartment model featuring first-order absorption and elimination. This model is consistent with predictable and dose-proportional pharmacokinetics. Indeed, systemic exposure to brivanib, as measured by the area under the concentration-time curve (AUC), was found to increase linearly with doses up to 1000 mg per day. The PPK modeling identified body weight as a statistically significant covariate affecting drug clearance, with clearance increasing as body weight increases. Other demographic factors, such as age, gender, and race, were not found to have a statistically significant impact on brivanib clearance.
Pharmacodynamic (PD) studies were conducted to link drug exposure to both biological effects and clinical toxicities, providing crucial insights into the drug's therapeutic window.
The clinical pharmacology profile of brivanib reveals a critical challenge that likely contributed to its ultimate failure. While the selection of the 800 mg QD dosing regimen was a rational attempt to optimize the therapeutic window by minimizing the -driven toxicity of fatigue, the results of the Phase III trials suggest this window remained too narrow. Despite this optimization, fatigue remained a prominent Grade 3/4 adverse event, and the high rate of treatment discontinuation due to toxicity indicates that even the "optimized" dose was poorly tolerated by a substantial portion of the patient population. This suggests that the plasma concentrations required to achieve meaningful, sustained target inhibition were inextricably linked to concentrations that caused dose-limiting side effects, a fundamental challenge that could not be overcome through dosing schedule modifications alone.
The clinical development of brivanib alaninate was extensive and ambitious, reflecting the initial optimism surrounding its dual-targeting mechanism. Bristol-Myers Squibb sponsored a global program that ultimately involved at least 29 clinical trials and enrolled more than 4,000 patients with various cancer types. While the strategic cornerstone of the program was advanced Hepatocellular Carcinoma (HCC), investigations were also conducted in several other solid tumors. These included studies in metastatic colorectal cancer, often evaluating brivanib in combination with the EGFR inhibitor cetuximab (e.g., NCT00640471) ; recurrent or persistent cervical cancer (NCT01267253) ; recurrent endometrial cancer (NCT00888173) ; and metastatic renal cell carcinoma (NCT01253668). However, the definitive assessment of brivanib's clinical utility hinged on the outcomes of the large-scale Phase III program in HCC, known as the BRISK (Brivanib studies in HCC patients at RISK) trials.
The BRISK-FL (First Line) study was designed to establish brivanib's role as a primary treatment for advanced HCC by comparing it directly against the established global standard of care, sorafenib.
Table 3: Efficacy Outcomes from the Phase III BRISK-FL Trial (Brivanib vs. Sorafenib)
| Endpoint | Brivanib (n=577) | Sorafenib (n=578) | Hazard Ratio (95.8% CI) | Comment |
|---|---|---|---|---|
| Median Overall Survival (OS) | 9.5 months | 9.9 months | 1.06 (0.93 - 1.22) | Primary endpoint of non-inferiority not met (upper CI > 1.08) |
| Time to Progression (TTP) | Similar | Similar | Not Reported | No significant difference observed |
| Objective Response Rate (ORR) | Similar | Similar | Not Reported | No significant difference observed |
| Disease Control Rate (DCR) | Similar | Similar | Not Reported | No significant difference observed |
| Data sourced from. |
The BRISK-PS (Post Sorafenib) study was designed to address a significant unmet medical need: effective treatment for HCC patients whose disease has progressed despite, or who are intolerant to, first-line sorafenib therapy.
Table 4: Efficacy Outcomes from the Phase III BRISK-PS Trial (Brivanib vs. Placebo)
| Endpoint | Brivanib + BSC (n=263) | Placebo + BSC (n=132) | Hazard/Odds Ratio (95% CI) | P-value |
|---|---|---|---|---|
| Median Overall Survival (OS) | 9.4 months | 8.2 months | 0.89 (0.69 - 1.15) | 0.3307 |
| Median Time to Progression (TTP) | 4.2 months | 2.7 months | 0.56 (0.42 - 0.76) | < 0.001 |
| Objective Response Rate (ORR, mRECIST) | 10% | 2% | 5.72 (Odds Ratio) | Not Reported |
| Data sourced from. Sample sizes reflect the 2:1 randomization. |
The collective results from the BRISK program paint a clear picture of a drug with demonstrable biological activity that ultimately failed to translate into a clinically meaningful survival benefit. The discordance observed in the BRISK-PS trial—where brivanib significantly delayed tumor progression and induced tumor shrinkage but did not help patients live longer—is a classic example of a drug with primarily cytostatic (growth-inhibiting) rather than cytotoxic (cell-killing) effects. This modest effect, while statistically significant, was likely insufficient in magnitude or duration to alter the overall trajectory of a highly aggressive disease like advanced HCC.
Furthermore, the failure in the BRISK-FL trial was likely driven by an inferior therapeutic index compared to sorafenib. While the drugs showed similar antitumor activity on secondary measures, the significantly poorer tolerability of brivanib, as evidenced by its higher discontinuation rate, meant that patients were less able to remain on therapy long enough to derive a potential survival benefit. In oncology, the ability of a patient to tolerate and adhere to a treatment regimen is a critical determinant of long-term outcomes. Brivanib's toxicity profile created a barrier to achieving the sustained drug exposure necessary for efficacy, a fatal flaw in a head-to-head comparison with an established, albeit imperfect, standard of care.
The safety and tolerability of brivanib alaninate were extensively evaluated throughout its clinical development, from early-phase dose-escalation studies to the large pivotal trials. Across Phase I and II studies, the toxicity profile was generally characterized as manageable, allowing for the determination of a Maximum Tolerated Dose (MTD) of 800 mg once daily, which was carried forward into Phase III development.
The most frequently reported treatment-related adverse events (AEs) of any severity included fatigue, hypertension, diarrhea, nausea, decreased appetite, and hyponatremia (low sodium levels). The overall pattern of toxicity was consistent with the known class effects of agents that inhibit the VEGF signaling pathway, with events like hypertension and proteinuria being common. More serious class-related toxicities, such as thromboembolic events and bleeding, were also observed, though less frequently. The compound's hazard profile, based on aggregated GHS (Globally Harmonized System) information, includes warnings that it is harmful if swallowed and may pose a risk of reproductive toxicity and organ damage with prolonged or repeated exposure.
The head-to-head comparison in the BRISK-FL trial and the placebo-controlled design of the BRISK-PS trial provided a definitive characterization of brivanib's safety profile and its relative tolerability. These trials revealed distinct and largely non-overlapping toxicity profiles between brivanib and sorafenib.
This comparative data suggests a critical qualitative difference in the nature of the toxicities. Sorafenib's primary dose-limiting toxicity is a localized dermatological condition, which, while significantly impacting quality of life, can often be managed symptomatically. Brivanib's primary toxicities, however, are systemic and metabolic in nature—fatigue, hypertension, and electrolyte disturbances. These types of systemic AEs can be more debilitating and may be perceived by both patients and clinicians as more serious, potentially leading to a lower threshold for discontinuing treatment.
Table 5: Comparative Incidence of Frequent Grade 3/4 Adverse Events in Pivotal HCC Trials
| Adverse Event | BRISK-FL: Brivanib (%) | BRISK-FL: Sorafenib (%) | BRISK-PS: Brivanib (%) | BRISK-PS: Placebo (%) |
|---|---|---|---|---|
| Hypertension | 13 | 5 | 17 | Not Reported |
| Fatigue | 15 | 7 | 13 | Not Reported |
| Hyponatremia | 23 | 9 | 11 | Not Reported |
| Hand-Foot-Skin Reaction | 2 | 15 | Not Reported | Not Reported |
| AST Elevation | 14 | 17 | Not Reported | Not Reported |
| Decreased Appetite | Not Reported | Not Reported | 10 | Not Reported |
| Diarrhea | Not Reported | Not Reported | Not Reported | Not Reported |
| Discontinuation due to AEs | 43 | 33 | 23 | 7 |
| Data sourced from. Not Reported indicates the AE was not listed among the most frequent in the source abstract. |
The most telling indicator of a drug's tolerability in a clinical setting is the rate at which patients must either reduce the dose or stop taking the drug altogether due to side effects. The data from the pivotal trials clearly demonstrate that brivanib was less well-tolerated than the standard of care.
In the BRISK-FL trial, a striking 43% of patients in the brivanib arm discontinued treatment due to adverse events, a rate significantly higher than the 33% observed in the sorafenib arm. This 10-point absolute difference is highly clinically meaningful and represents a major disadvantage for brivanib. Interestingly, the rates of dose reduction were nearly identical between the two arms (49% for brivanib vs. 50% for sorafenib). This suggests that for a substantial proportion of patients experiencing toxicity on brivanib, simply lowering the dose was not a sufficient management strategy, necessitating complete cessation of therapy.
The BRISK-PS trial further confirmed the significant toxicity burden of the drug. In this study, 23% of patients receiving brivanib discontinued due to AEs, more than three times the rate of 7% seen in the placebo arm. This high discontinuation rate, driven by a challenging safety profile, is a primary factor in explaining the drug's failure to demonstrate an overall survival benefit. A drug cannot help a patient live longer if its side effects prevent the patient from taking it for a sufficient duration.
In recognition of the significant unmet medical need in advanced hepatocellular carcinoma and based on promising early-phase clinical data, brivanib alaninate was granted Orphan Drug Designation by major regulatory agencies. This status is intended to facilitate the development of drugs for rare diseases. The European Medicines Agency (EMA), through the European Commission, granted orphan designation (EU/3/11/918) on October 27, 2011. The basis for this designation was preliminary evidence suggesting that brivanib might offer a significant benefit for HCC patients, particularly as an alternative for those who had failed or could not tolerate existing treatments. Similarly, the U.S. Food and Drug Administration (FDA) granted orphan designation for the treatment of HCC on March 18, 2011.
However, these designations are contingent on the continued demonstration of a favorable risk-benefit profile. Following the definitive negative results from the pivotal Phase III clinical trial program, these designations were withdrawn. The FDA designation was officially withdrawn on June 27, 2014, and the EMA designation was also withdrawn, marking the formal end of its privileged regulatory status.
The outcomes of the BRISK-FL and BRISK-PS trials were the decisive factors in the fate of brivanib alaninate. Following the announcement in early 2012 that the BRISK-FL study had failed to meet its primary endpoint of non-inferiority to sorafenib, Bristol-Myers Squibb stated that it was "considering options for the ongoing brivanib development program". The subsequent failure of the BRISK-PS trial to demonstrate a survival benefit over placebo solidified the conclusion that the drug did not have a viable path forward in HCC. Consequently, Bristol-Myers Squibb officially discontinued the development of brivanib. This decision was a direct and responsible response to the high-quality, unambiguous evidence generated by the large-scale Phase III program, which demonstrated that the drug did not offer a meaningful clinical benefit to patients with advanced HCC.
Brivanib alaninate stands as a significant and illustrative case study from a challenging era in the development of therapies for HCC, roughly spanning from 2010 to 2015. After the landmark approval of sorafenib, the first systemic agent to show a survival benefit in HCC, the field saw a wave of new tyrosine kinase inhibitors (TKIs) enter late-stage development, many of which, including sunitinib and brivanib, ultimately failed to demonstrate superiority or even non-inferiority to the new standard of care.
The story of brivanib's development and discontinuation underscores several enduring principles in modern oncology drug development:
In conclusion, the development of brivanib alaninate was a scientifically rigorous and well-executed program that asked a valid and important clinical question. The definitive, albeit negative, answer provided by the Phase III trials contributed valuable knowledge to the field and highlighted the immense difficulty of improving upon the therapeutic index of multi-targeted kinase inhibitors in a complex and heterogeneous disease like hepatocellular carcinoma.
Published at: October 11, 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.