MedPath

Vorasidenib Advanced Drug Monograph

Published:Aug 28, 2025

Generic Name

Vorasidenib

Drug Type

Small Molecule

Chemical Formula

C14H13ClF6N6

CAS Number

1644545-52-7

Vorasidenib (Voranigo): A Comprehensive Monograph on the First-in-Class Targeted Therapy for IDH-Mutant Glioma

Executive Summary

Vorasidenib, marketed under the brand name Voranigo, represents a landmark achievement in the field of neuro-oncology and a paradigm shift in the management of Grade 2 diffuse gliomas harboring mutations in the isocitrate dehydrogenase 1 or 2 genes (IDH1/2). As a first-in-class, orally available, brain-penetrant dual inhibitor of mutant IDH1 and IDH2 enzymes, vorasidenib directly targets the fundamental oncogenic driver of these tumors. The development of this agent was predicated on a deep understanding of the molecular pathogenesis of IDH-mutant gliomas, specifically the role of the oncometabolite D-2-hydroxyglutarate (2-HG) in disrupting epigenetic regulation and blocking cellular differentiation. Vorasidenib was rationally designed to overcome the critical limitation of previous IDH inhibitors—poor blood-brain barrier penetrance—thereby enabling effective therapeutic concentrations at the tumor site.

The clinical value of vorasidenib was unequivocally established in the pivotal Phase 3 INDIGO trial, a global, randomized, placebo-controlled study. The trial demonstrated statistically unprecedented and clinically transformative efficacy, more than doubling the median progression-free survival (PFS) from 11.1 months with placebo to 27.7 months with vorasidenib. Even more profoundly, it delayed the median time to next intervention (TTNI)—the need for subsequent cytotoxic radiation or chemotherapy—from 17.8 months to a duration not yet reached in the treatment arm, representing a 74% reduction in risk. These results were achieved with a manageable and favorable safety profile, with the most significant adverse event being reversible transaminase elevations, which can be effectively managed with routine monitoring.

Vorasidenib’s compelling efficacy and safety data, coupled with the significant unmet need in this patient population, led to a rapid and harmonized global regulatory approval process. It received a full suite of expedited designations from the U.S. Food and Drug Administration (FDA), culminating in its approval on August 6, 2024, as the first-ever targeted therapy for Grade 2 IDH-mutant glioma. Similar accelerated assessments and approvals have followed from the European Medicines Agency (EMA) and other international regulatory bodies. Vorasidenib has redefined the standard of care, transitioning the treatment paradigm from a reactive "watch-and-wait" approach or the premature use of toxic therapies to a proactive, targeted, and well-tolerated intervention. By providing a means to control tumor growth and significantly delay the onset of debilitating treatments, vorasidenib preserves neurological function and quality of life, establishing a new therapeutic cornerstone for patients with this chronic and life-altering malignancy.

Introduction: The Unmet Need in Grade 2 IDH-Mutant Glioma

Characterization of Grade 2 Diffuse Gliomas

Grade 2 diffuse gliomas, encompassing astrocytomas and oligodendrogliomas, are malignant primary brain tumors that, despite their "low-grade" designation, follow an inexorably progressive clinical course.[1] These tumors typically afflict a younger patient population, with a median age at diagnosis of approximately 40 years, an age when individuals are often at the peak of their professional and personal lives.[3] Although initially slow-growing, these neoplasms are diffusely infiltrative and cannot be cured by surgery alone.[1] Over time, they invariably recur and often undergo malignant transformation to higher-grade tumors, leading to progressive neurological disability, cognitive decline, and ultimately, premature mortality.[2] The deceptive indolence of the early stages of the disease belies its lethal nature, creating a profound and long-standing challenge for both patients and clinicians.

The Central Role of IDH Mutations

A seminal discovery in the last two decades that revolutionized the understanding and classification of these tumors was the identification of recurrent, somatic, gain-of-function mutations in the genes encoding the metabolic enzymes isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2).[2] These mutations are a disease-defining molecular hallmark, present in approximately 80% of Grade 2 gliomas and a smaller fraction of Grade 3 gliomas.[2] IDH mutations are considered a truncal, or foundational, oncogenic event, occurring early in gliomagenesis and remaining stable throughout the disease course.[2]

Unlike the wild-type enzymes, which catalyze the conversion of isocitrate to alpha-ketoglutarate (α-KG) within the Krebs cycle, the mutant IDH1/2 enzymes acquire a neomorphic catalytic activity. They convert α-KG into the oncometabolite D-2-hydroxyglutarate (2-HG).[2] Pathologically high concentrations of 2-HG competitively inhibit

α-KG-dependent dioxygenases, including histone and DNA demethylases. This leads to widespread epigenetic dysregulation, most notably a global DNA hypermethylation phenotype, which blocks normal cellular differentiation and promotes a state primed for oncogenesis.[5] This detailed molecular understanding provided a clear and compelling scientific rationale: targeting the mutant IDH enzymes to block 2-HG production could represent a highly specific and effective therapeutic strategy to halt the primary driver of the disease.

The Paradox of the Traditional Standard of Care

Prior to the advent of targeted therapy, the management of Grade 2 IDH-mutant glioma was defined by a difficult and unsatisfactory clinical dilemma. The standard of care (SoC) began with maximal safe surgical resection to establish a diagnosis and reduce tumor burden.[1] Following surgery, patients and their physicians faced a challenging choice with no clear optimal path.

One option was a period of active surveillance, often termed "watch-and-wait," where no further treatment was administered until there was definitive evidence of radiographic or clinical progression.[4] While this approach deferred the toxicity of further treatment, it came at the cost of significant psychological burden for patients, who were forced to live with the knowledge of an incurable tumor growing in their brain.[5]

The alternative was to proceed with adjuvant therapy, typically radiation (XRT) with or without chemotherapy, such as the procarbazine, lomustine, and vincristine (PCV) regimen or temozolomide.[1] While clinical trials demonstrated that adjuvant chemoradiotherapy could improve progression-free survival, this benefit was achieved at the expense of substantial and often irreversible long-term toxicities.[1] For a young patient population with a potential life expectancy of one to two decades, the consequences of these treatments—including profound neurocognitive impairment, memory loss, executive dysfunction, radiation-induced necrosis, leukoencephalopathy, and an increased risk of secondary malignancies—were devastating and severely impacted quality of life.[2]

Defining the Therapeutic Gap

This stark choice between anxious observation and proactive but debilitating treatment created a profound therapeutic gap. There was a desperate unmet need for an intervention that could be administered early in the disease course, following surgery, to effectively control tumor growth without inflicting the collateral damage of conventional cytotoxic therapies.[2] The development of vorasidenib was a direct and rational response to this need. The goal was to create a targeted, well-tolerated therapy that could fundamentally alter the natural history of the disease by delaying or even preventing the need for radiation and chemotherapy, thereby preserving long-term neurological function and quality of life for patients.

The approval of vorasidenib does not merely represent the addition of a new drug to the treatment algorithm; it signifies a fundamental philosophical shift in the management of low-grade glioma. The historical paradigm forced a reactive posture, where the inevitable progression of the tumor would trigger the deployment of toxic therapies. Vorasidenib's success validates a new, proactive strategy: the early use of a targeted agent to control the underlying biology of the disease. This approach transforms the "watch-and-wait" period into a "watch-and-treat" period, empowering patients and clinicians with an effective tool to manage the disease from an earlier stage, fundamentally changing the conversation from when to start harsh treatments to how long they can be safely and effectively delayed.

Molecular Profile and Physicochemical Properties

Chemical Identity and Structure

Vorasidenib is a small molecule, orally bioavailable antineoplastic agent belonging to the triazine class of compounds.[8] Its systematic International Union of Pure and Applied Chemistry (IUPAC) name is 6-(6-chloropyridin-2-yl)-2-N,4-N-bis-1,3,5-triazine-2,4-diamine.[8] The chemical structure is characterized by a central 1,3,5-triazine ring. This core is substituted with a 6-chloropyridin-2-yl group at one position and two identical chiral (2R)-1,1,1-trifluoropropan-2-ylamino side chains at the other two positions.[10] This specific molecular architecture is the result of extensive medicinal chemistry optimization aimed at achieving potent dual inhibition of mutant IDH1/2 enzymes while conferring the critical property of blood-brain barrier penetration.[14]

The development of vorasidenib was a direct response to the failures of its predecessors. The drug's developer had previously created ivosidenib (a mutant IDH1 inhibitor) and enasidenib (a mutant IDH2 inhibitor), which received approval for hematologic malignancies.[8] However, these earlier agents exhibited poor brain exposure, rendering them ineffective for treating gliomas, the very tumors where IDH mutations are most prevalent.[8] The molecular design of vorasidenib, therefore, was not accidental but a deliberate effort to solve this central problem. Its physicochemical properties were carefully tuned to enhance its ability to cross the blood-brain barrier, a feat its forerunners could not achieve. Furthermore, its design as a dual inhibitor of both mIDH1 and mIDH2 was a strategic choice to provide broader coverage and potentially circumvent resistance mechanisms such as isoform switching.[8]

Physicochemical Properties and Identifiers

The fundamental chemical and physical identifiers for vorasidenib are consolidated in Table 3.1. These data provide a standardized reference for the molecule's identity across scientific and regulatory databases.

Table 3.1: Key Chemical and Physical Identifiers of Vorasidenib

PropertyValueSource(s)
Drug NameVorasidenib8
Brand NameVoranigo8
DrugBank IDDB170978
CAS Number1644545-52-78
TypeSmall Molecule11
Molecular FormulaC14​H13​ClF6​N6​8
Molar Mass414.74 g/mol8
IUPAC Name6-(6-chloropyridin-2-yl)-2-N,4-N-bis-1,3,5-triazine-2,4-diamine8
InChIKeyQCZAWDGAVJMPTA-RNFRBKRXSA-N8
Canonical SMILESCC@HNC1=NC(=NC(=N1)C2=NC(=CC=C2)Cl)NC(F)(F)F
SynonymsAG-881, AG881

Calculated Properties and Drug-Likeness

Computational analysis of vorasidenib's structure reveals physicochemical properties that are highly favorable for an orally administered drug targeting the central nervous system (CNS). Its calculated partition coefficient (AlogP) is 4.31, indicating a high degree of lipophilicity, which is a key factor in facilitating passive diffusion across the blood-brain barrier. The molecule has a topological polar surface area (TPSA) of 75.62

A˚2, two hydrogen bond donors, and six hydrogen bond acceptors.

These parameters are consistent with established guidelines for oral bioavailability and CNS penetration. Notably, vorasidenib adheres to Lipinski's Rule of Five, with zero violations reported. This rule is a widely used heuristic in drug discovery to predict the "drug-likeness" of a chemical compound, and adherence suggests a high probability of good membrane permeability and oral absorption. The combination of these properties underscores the successful rational design of vorasidenib as a brain-penetrant agent, directly addressing the critical shortcoming of earlier-generation IDH inhibitors.

Clinical Pharmacology

4.1. Mechanism of Action (MOA)

Dual Inhibition of Mutant IDH1/IDH2

Vorasidenib functions as a potent, reversible, first-in-class dual inhibitor of the mutated forms of both isocitrate dehydrogenase 1 (IDH1), located in the cytoplasm and peroxisomes, and isocitrate dehydrogenase 2 (IDH2), located in the mitochondria. It exhibits broad activity against a range of clinically relevant IDH mutations, including IDH1 R132H, R132C, R132G, and R132S, as well as IDH2 R140Q and R172K. X-ray crystallography studies have elucidated that vorasidenib binds to an allosteric site at the interface of the enzyme dimer, locking it in an inactive, open conformation that prevents substrate binding and catalytic activity.

Suppression of Oncometabolite 2-HG

The pathogenic hallmark of IDH-mutant gliomas is the massive accumulation of the oncometabolite D-2-hydroxyglutarate (2-HG), which can reach millimolar concentrations within tumor cells. This accumulation is a direct consequence of the neomorphic activity of the mutant IDH1/2 enzymes. As described previously, 2-HG acts as a competitive inhibitor of numerous

α-KG-dependent dioxygenases, leading to profound epigenetic alterations and a block in cellular differentiation that are central to gliomagenesis.

Therapeutic Effect

The primary therapeutic mechanism of vorasidenib is the potent and specific inhibition of mutant IDH1 and IDH2 enzymes, which directly halts the aberrant production of 2-HG. Clinical evidence from a surgical substudy has confirmed that administration of vorasidenib leads to a substantial reduction in 2-HG levels within the tumor tissue. By lowering intratumoral 2-HG concentrations, vorasidenib is believed to reverse the 2-HG-mediated epigenetic blockade. This allows for the restoration of normal histone and DNA methylation patterns, ultimately leading to the differentiation of malignant glioma cells and a concomitant reduction in their proliferation. This targeted cytostatic effect, which promotes differentiation rather than immediate cell death, is consistent with the observed clinical outcome of slowed tumor growth and prolonged disease stability.

4.2. Pharmacokinetics (ADME Profile)

The pharmacokinetic (PK) profile of vorasidenib is characterized by properties that are exceptionally well-suited for its intended clinical use in treating a chronic CNS malignancy. The synergy between its efficient CNS delivery, convenient dosing schedule, and high target specificity creates a highly favorable therapeutic index.

Absorption

Vorasidenib is administered orally. At steady state, the median time to reach maximum plasma concentration (Tmax) is 2 hours, with a range of 0.5 to 4 hours. The drug's absorption is significantly influenced by food. A high-fat, high-calorie meal increases the maximum concentration (Cmax) by 3.1-fold and the total exposure (Area Under the Curve, AUC) by 1.4-fold compared to fasting conditions. This food effect necessitates consistent administration relative to meals, though the prescribing information allows for dosing with or without food. The mean absolute bioavailability is 34%.

Distribution

Vorasidenib exhibits extensive distribution into tissues, as indicated by its large mean steady-state volume of distribution (Vd​) of 3,930 L. A critical feature of its distribution profile is its ability to effectively penetrate the blood-brain barrier (BBB), a non-negotiable requirement for any CNS-targeted therapy. Studies have demonstrated a brain tumor-to-plasma concentration ratio of 1.6, confirming that therapeutically relevant concentrations are achieved at the site of action. In the bloodstream, vorasidenib is highly bound to plasma proteins (97%).

Metabolism

The primary metabolic pathway for vorasidenib is oxidation mediated by the cytochrome P450 enzyme system, with CYP1A2 being the major contributor. Minor contributions are made by several other isoforms, including CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A. This reliance on CYP1A2 for clearance is the basis for clinically significant drug-drug interaction warnings, particularly with strong inhibitors or inducers of this enzyme. Vorasidenib itself also acts as an inducer of CYP3A, which can affect the metabolism of co-administered drugs that are substrates of that enzyme.

Excretion

Vorasidenib and its metabolites are eliminated predominantly through the fecal route. Following a single radiolabeled oral dose, 85% of the radioactivity was recovered in the feces, with 56% of that being the unchanged parent drug. Renal excretion is a minor pathway, accounting for only 4.5% of the dose recovered in urine. The drug has a notably long mean terminal elimination half-life (

t1/2​) of 10 days at steady state. This long half-life is highly advantageous, as it supports a convenient once-daily dosing regimen, which is crucial for maintaining adherence and quality of life during long-term, chronic therapy.

Table 4.1: Summary of Key Pharmacokinetic Parameters of Vorasidenib

ParameterValueSource(s)
Tmax (Time to Peak Concentration)2 hours (median)
Bioavailability (Absolute)34%
Volume of Distribution (Vd​)3,930 L
Plasma Protein Binding97%
Brain Tumor-to-Plasma Ratio1.6
Primary Metabolism PathwayCYP1A2
Terminal Half-Life (t1/2​)10 days
Route of EliminationPrimarily fecal (85%)

Clinical Efficacy and Development

The clinical development program for vorasidenib stands as a model of efficient, hypothesis-driven research. Insights gained from the early-phase trial, which identified the specific patient population most likely to benefit, were astutely applied to the design of the pivotal Phase 3 study. This precise targeting maximized the probability of success and resulted in one of the most statistically robust and clinically meaningful outcomes in the history of neuro-oncology.

5.1. Early Phase Development (First-in-Human Trial NCT02481154)

Study Design and Population

The first-in-human evaluation of vorasidenib was a multicenter, open-label, Phase 1 dose-escalation and expansion study (NCT02481154). The trial enrolled 93 patients with advanced solid tumors harboring an IDH1 or IDH2 mutation, which included a specific cohort of 52 patients with recurrent or progressive glioma who had previously undergone standard therapy. The primary objectives were to assess the safety, tolerability, pharmacokinetics, and to determine the maximum tolerated dose (MTD) and recommended Phase 2 dose.

Safety and Dose Finding

The study established a favorable and manageable safety profile for vorasidenib. Dose-limiting toxicities (DLTs) were identified as reversible, dose-dependent elevations in liver transaminases (ALT/AST), which occurred at dose levels of 100 mg once daily and higher. These elevations resolved with dose interruption or reduction. Based on these findings, a dose of 50 mg once daily was initially selected for further study in glioma, which was subsequently refined to the 40 mg daily dose used in the pivotal Phase 3 trial.

Pivotal Efficacy Signal

The Phase 1 trial provided the first crucial evidence of vorasidenib's antitumor activity in glioma and, critically, generated a powerful hypothesis regarding the optimal patient population for treatment. A stark divergence in clinical benefit was observed based on the radiological features of the tumors.

For the subgroup of patients with non-enhancing glioma (n=22)—tumors that are radiologically consistent with a lower-grade, less aggressive biology—vorasidenib demonstrated encouraging and durable clinical activity. The objective response rate was 18% (including one partial response and three minor responses), and the median progression-free survival (PFS) was an impressive 36.8 months.

In stark contrast, for patients with enhancing glioma (n=28)—a feature that often signifies progression to a higher grade and more aggressive behavior—the drug showed minimal benefit. There were no objective responses, and the median PFS was only 3.6 months. This critical observation suggested that while vorasidenib is highly effective against the primary oncogenic driver in lower-grade disease, tumors that have progressed to an enhancing state may have acquired additional genetic alterations or activated alternative signaling pathways, rendering them less dependent on the IDH mutation for survival and growth. This insight was not overlooked; it became the foundational principle upon which the successful Phase 3 trial was built.

5.2. The Pivotal Phase 3 INDIGO Trial (NCT04164901)

Study Rationale and Design

Informed by the clear efficacy signal in the non-enhancing glioma cohort from the Phase 1 study, the INDIGO trial (NCT04164901) was designed to definitively test vorasidenib in this specific, well-defined patient population. It was a global, randomized, double-blind, placebo-controlled Phase 3 study, the gold standard for clinical evidence. The trial enrolled 331 patients, aged 12 years or older, with residual or recurrent Grade 2 astrocytoma or oligodendroglioma with a confirmed IDH1 or IDH2 mutation. A key eligibility criterion was that patients had undergone surgery one to five years prior but had received no other anticancer treatment, placing them squarely in the "watch-and-wait" period where the greatest therapeutic need existed. Patients were randomized on a 1:1 basis to receive either vorasidenib 40 mg orally once daily or a matching placebo.

Primary Endpoint: Progression-Free Survival (PFS)

The INDIGO trial met its primary endpoint with a level of statistical significance and clinical benefit rarely seen in oncology trials. At the second planned interim analysis, the study was unblinded early due to overwhelming evidence of efficacy. Vorasidenib treatment resulted in a median PFS of 27.7 months, as assessed by a blinded independent radiology committee, compared to just 11.1 months for patients in the placebo group. This represented a statistically significant and clinically profound improvement, with a hazard ratio (HR) for progression or death of 0.39 (95% Confidence Interval [CI]: 0.27 to 0.56;

p<0.0001). This translates to a 61% reduction in the risk of disease progression or death for patients treated with vorasidenib.

Key Secondary Endpoint: Time to Next Intervention (TTNI)

Equally, if not more, important from a patient perspective was the key secondary endpoint of time to next intervention (TTNI), which measures the time until a patient requires subsequent treatment, such as radiation or chemotherapy. Vorasidenib demonstrated an even more dramatic benefit on this endpoint. The median TTNI for patients receiving placebo was 17.8 months. For patients receiving vorasidenib, the median TTNI had not yet been reached at the time of analysis, indicating a very substantial delay. The hazard ratio was 0.26 (95% CI: 0.15 to 0.43;

p<0.0001), corresponding to a 74% reduction in the risk of needing to start the next line of toxic therapy. This result directly validated the core therapeutic goal of vorasidenib: to safely and effectively postpone the need for treatments that carry a heavy burden of long-term toxicity.

Other Efficacy Measures

In addition to delaying progression, vorasidenib was shown to actively induce tumor regression. Volumetric MRI analysis revealed that tumors in patients treated with vorasidenib shrank by a mean of 2.5% every six months. In contrast, tumors in the placebo arm continued to grow, increasing in volume by a mean of 13.9% over the same period. This demonstrated that vorasidenib provides not just disease stabilization but an active, durable antitumor effect.

Table 5.1: Summary of Primary and Key Secondary Efficacy Endpoints from the INDIGO Trial

Data sourced from

Patient Management and Clinical Use

6.1. Approved Indication and Patient Selection

Indication

Vorasidenib is indicated for the treatment of adult and pediatric patients aged 12 years and older with Grade 2 astrocytoma or oligodendroglioma that possesses a susceptible isocitrate dehydrogenase-1 (IDH1) or isocitrate dehydrogenase-2 (IDH2) mutation. The indication specifies its use in the post-surgical setting, which can include patients who have undergone biopsy, sub-total resection, or gross total resection of their tumor.

Patient Selection

A critical prerequisite for initiating therapy is the confirmation of a susceptible IDH1 or IDH2 mutation within the patient's tumor tissue. This molecular diagnosis must be established using a validated, FDA-approved diagnostic test. This requirement anchors the use of vorasidenib firmly in the principles of precision oncology, ensuring that the treatment is administered only to those patients whose tumors harbor the specific molecular target of the drug.

6.2. Dosing, Administration, and Monitoring Protocols

Dosing

The recommended dosage of vorasidenib is weight-based for the pediatric population and fixed for adults.

  • Adult patients and pediatric patients (12 years and older) weighing ≥40 kg: 40 mg administered orally once daily.
  • Pediatric patients (12 years and older) weighing <40 kg: 20 mg administered orally once daily.

Treatment should be continued until evidence of disease progression or the development of unacceptable toxicity.

Administration

Vorasidenib tablets are to be swallowed whole and should not be split, crushed, or chewed. The medication can be taken with or without food but should be administered at approximately the same time each day to maintain consistent plasma concentrations. If a dose is missed by more than 6 hours, the patient should skip that dose and take the next one at the regularly scheduled time. If vomiting occurs after a dose, a replacement dose should not be taken; the patient should wait until the next scheduled dose.

Dose Modifications

The full prescribing information provides specific guidelines for dose modifications in the event of adverse reactions. For adults (and pediatrics ≥40 kg), the first dose reduction is to 20 mg daily, and the second is to 10 mg daily. For pediatric patients <40 kg, the first dose reduction is to 10 mg daily. If a patient is unable to tolerate the 10 mg daily dose, treatment with vorasidenib should be permanently discontinued.

Monitoring

Given the risk of hepatotoxicity, a strict monitoring protocol is mandatory.

  • Liver Function Tests (LFTs): Alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transferase (GGT), and total bilirubin must be assessed prior to starting treatment.
  • On-Treatment Monitoring: LFTs should be monitored every two weeks for the first two months of therapy, then monthly for the first two years, and as clinically indicated thereafter. More frequent monitoring is required for patients who develop transaminase elevations.
  • Pregnancy Status: Pregnancy status must be verified in females of reproductive potential before initiating treatment.

6.3. Comprehensive Safety and Tolerability Profile

Overview

Across the clinical development program, vorasidenib has demonstrated a manageable and generally favorable safety profile, particularly when compared to the significant toxicities associated with radiation and chemotherapy. In the pivotal INDIGO trial, the overall quality of life reported by patients in the vorasidenib arm was comparable to that of patients receiving placebo, indicating a low treatment burden.

Most Common Adverse Reactions

The most frequently reported adverse reactions (occurring in ≥15% of patients) in the INDIGO trial were fatigue, headache, COVID-19 infection, musculoskeletal pain, diarrhea, nausea, and seizure. The incidence of many of these events, such as fatigue and headache, was similar between the vorasidenib and placebo arms, suggesting a contribution from the underlying disease or other factors.

Warnings and Precautions - Hepatotoxicity

The most clinically significant safety concern associated with vorasidenib is the risk of hepatotoxicity. Elevations in liver transaminases are common. In the INDIGO trial, Grade 3 or 4 elevations in ALT occurred in 10% of patients and Grade 3 or 4 elevations in AST occurred in 4.8% of patients. These events are typically reversible with dose modification or interruption. However, rare but serious events, including cases meeting Hy’s Law criteria, autoimmune hepatitis, and hepatic failure, have been reported, necessitating the stringent monitoring protocol outlined above.

Warnings and Precautions - Embryo-Fetal Toxicity

Based on findings from animal reproduction studies, vorasidenib can cause harm to a developing fetus. Consequently, it is crucial to advise patients of this potential risk. Females of reproductive potential must use effective nonhormonal contraception during treatment and for three months following the final dose, as vorasidenib can reduce the efficacy of hormonal contraceptives. Male patients with female partners of reproductive potential should also use effective contraception during the same period.

Table 6.1: Adverse Reactions Reported in the INDIGO Trial (Vorasidenib vs. Placebo)

Incidence based on laboratory abnormalities, worsened from baseline. Data sourced from.

6.4. Drug Interactions and Special Populations

CYP1A2 Interactions

As vorasidenib is primarily metabolized by CYP1A2, co-administration with drugs that strongly or moderately inhibit this enzyme (e.g., ciprofloxacin, fluvoxamine) should be avoided, as this can significantly increase vorasidenib exposure and the risk of toxicity. Conversely, co-administration with moderate or strong CYP1A2 inducers (e.g., carbamazepine, rifampin), including tobacco smoke, should be avoided as this can decrease vorasidenib exposure and potentially compromise its efficacy.

CYP3A Interactions

Vorasidenib is an inducer of the CYP3A enzyme family. Therefore, caution is advised when co-administering vorasidenib with CYP3A substrates, particularly those with a narrow therapeutic index (e.g., certain immunosuppressants or chemotherapeutic agents), as vorasidenib may reduce their plasma concentrations and efficacy.

Hormonal Contraceptives

Due to its CYP3A induction properties, vorasidenib can decrease the plasma concentrations of hormonal contraceptives, potentially leading to contraceptive failure. As noted, females of reproductive potential who are taking vorasidenib must be counseled to use effective nonhormonal methods of contraception.

Fertility

Animal studies have indicated that vorasidenib may impair fertility in both males and females. In rat studies, these effects were not reversible. Patients of reproductive potential should be counseled on this risk.

Regulatory Trajectory and Market Access

The regulatory journey of vorasidenib was characterized by remarkable speed and global harmonization, reflecting a worldwide consensus among health authorities on the drug's profound clinical benefit and the urgent unmet need it addresses. This process serves as an exemplary case study in how regulatory agencies can collaborate effectively to accelerate patient access to a transformative therapy for a rare and serious disease.

U.S. Food and Drug Administration (FDA) Approval

Vorasidenib, developed by Servier Pharmaceuticals under the brand name Voranigo, received approval from the U.S. FDA on August 6, 2024. This was a historic decision, as it marked the first approval of any systemic therapy specifically for the treatment of patients with Grade 2 astrocytoma or oligodendroglioma with a susceptible IDH1 or IDH2 mutation.

The significance of the drug was recognized early in its development, and the FDA granted its New Drug Application (NDA) a full complement of expedited program designations:

  • Fast Track Designation: To facilitate development and expedite the review of drugs to treat serious conditions and fill an unmet medical need.
  • Breakthrough Therapy Designation: Granted to drugs that are intended to treat a serious condition and where preliminary clinical evidence indicates substantial improvement over available therapy on a clinically significant endpoint.
  • Priority Review: Which directs overall attention and resources to the evaluation of applications for drugs that, if approved, would be significant improvements in the safety or effectiveness of the treatment of serious conditions.
  • Orphan Drug Designation: Which provides incentives to assist and encourage the development of drugs for rare diseases.

The combination of these designations, awarded in response to the unambiguous and highly compelling data from the INDIGO trial, ensured a swift and efficient review process, culminating in an approval ahead of the scheduled PDUFA action date.

European Medicines Agency (EMA) and Global Status

The regulatory momentum for vorasidenib extended globally. In Europe, the European Medicines Agency (EMA) granted the marketing authorization application an accelerated assessment, a pathway reserved for medicines of major public health interest and therapeutic innovation. Following this expedited review, the EMA's Committee for Medicinal Products for Human Use (CHMP) adopted a positive opinion on July 24, 2025, formally recommending the granting of a marketing authorization for Voranigo in the European Union.

A key factor in this rapid global rollout was the utilization of the FDA's Project Orbis initiative. This framework allows for the concurrent submission and collaborative review of oncology drug applications among international regulatory partners. For vorasidenib, the FDA collaborated with agencies in Australia (TGA), Brazil (ANVISA), Canada (Health Canada), and Switzerland (Swissmedic), among others. This parallel review process, facilitated by the strength of the INDIGO data, led to a cascade of near-simultaneous approvals in multiple major markets worldwide, ensuring that patients across the globe could gain access to this breakthrough therapy in a timely manner.

Market Access

Reflecting its status as a specialized, high-value oncology therapeutic, vorasidenib is being made available to patients through a limited distribution model. In the United States, Servier has partnered with specialty pharmacies, such as Biologics by McKesson, to manage the distribution of Voranigo. This approach ensures that patients receive the necessary high-touch support, including education on administration and side effect management, assistance with insurance and financial counseling, and coordination of care with their neuro-oncology team.

Strategic Analysis and Future Perspectives

8.1. Redefining the Standard of Care

Paradigm Shift

The approval of vorasidenib is not an incremental advance; it is a fundamental disruption of a treatment paradigm that had remained stagnant for over two decades. For the first time, clinicians have a targeted, non-cytotoxic tool to proactively manage Grade 2 IDH-mutant glioma in the crucial post-surgical period. This establishes a new standard of care, transforming the "watch-and-wait" phase into an active treatment phase with a well-tolerated oral therapy. This shift moves the field away from a reactive model, where treatment was dictated by tumor progression, to a proactive one, where the underlying biology of the tumor is controlled from an earlier stage.

Clinical Impact

The most immediate and profound clinical impact of vorasidenib is the significant delay in the need for subsequent radiation and chemotherapy. By pushing back the timeline for these toxic interventions, vorasidenib allows patients, who are typically in the prime of their lives, to live longer without the debilitating side effects that compromise neurocognitive function, professional capacity, and overall quality of life. This directly resolves the central therapeutic dilemma that previously defined the management of this disease. The focus of care can now shift from a difficult trade-off between tumor control and treatment toxicity to the preservation of long-term health and function, a truly patient-centric outcome.

8.2. Future Research Directions

The success of vorasidenib has invigorated the field of neuro-oncology, serving as a powerful proof-of-concept for targeted therapy in low-grade gliomas and opening up numerous avenues for future investigation. Its approval is not an endpoint but rather a catalyst for a new wave of clinical and translational research.

Combination Therapies

With vorasidenib established as a safe and effective monotherapy, the logical next step is to explore its potential in combination with other treatment modalities. Several clinical trials are already underway or planned:

  • Immunotherapy: A Phase 1 study (NCT05484622) is actively evaluating the safety and preliminary efficacy of combining vorasidenib with the PD-1 inhibitor pembrolizumab in patients with recurrent or progressive enhancing IDH-mutant glioma. The rationale is that by altering the tumor microenvironment, vorasidenib may synergize with checkpoint inhibitors to elicit a more robust anti-tumor immune response.
  • Chemotherapy: Another active trial is investigating the combination of vorasidenib with the standard-of-care chemotherapy agent temozolomide, exploring whether a dual-pronged attack on the tumor could yield even greater benefit.

New Settings - Maintenance Therapy

The therapeutic role of vorasidenib may extend beyond the initial post-surgical setting. The VIGOR trial (NCT06809322) is a large, randomized Phase 3 study designed to evaluate vorasidenib as a maintenance therapy after patients with higher-risk Grade 2 or Grade 3 astrocytoma have completed standard first-line chemoradiotherapy. If positive, this trial could establish a role for vorasidenib across the entire continuum of care, from initial treatment to long-term maintenance, further delaying subsequent progression.

Unanswered Questions

Despite its success, critical long-term questions remain. The ultimate impact of vorasidenib on overall survival (OS) is not yet known and will require extended follow-up from the INDIGO trial. Understanding the mechanisms of eventual resistance to vorasidenib will be crucial for developing subsequent lines of therapy. Furthermore, its role in higher-grade (Grade 3) IDH-mutant gliomas, and whether it can be effectively integrated with standard radiotherapy and chemotherapy in that setting, are areas of active and future investigation.

Conclusion

Vorasidenib (Voranigo) represents a monumental advance in the field of neuro-oncology, culminating from decades of fundamental research that first identified the central role of IDH mutations in gliomagenesis. It is the embodiment of precision medicine, a therapy rationally designed to inhibit the core oncogenic driver of a specific subset of brain tumors. Its molecular structure was meticulously optimized to overcome the critical challenge of the blood-brain barrier, enabling it to reach its target in therapeutically effective concentrations.

The clinical evidence, spearheaded by the landmark Phase 3 INDIGO trial, is unequivocal. Vorasidenib provides a profound and statistically unprecedented benefit in prolonging progression-free survival and, most critically, in delaying the time to next intervention for patients with Grade 2 IDH-mutant glioma. It achieves this with a favorable safety profile that preserves patients' quality of life, a stark contrast to the debilitating toxicities of conventional radiation and chemotherapy.

Vorasidenib is not merely an incremental addition to the therapeutic armamentarium; it is a paradigm-shifting agent that has fundamentally redefined the standard of care. It transforms the management of this disease from a reactive to a proactive strategy, offering a well-tolerated oral therapy that can be deployed early to control tumor growth. For a patient population that has waited more than twenty years for a meaningful therapeutic breakthrough, vorasidenib offers not just extended time, but quality time. It stands as a cornerstone of modern, molecularly-guided neuro-oncology and serves as both a benchmark and a catalyst for future innovation in the treatment of brain tumors.

Works cited

  1. SEOM-GEINO clinical guidelines for grade 2 gliomas (2023) - PMC, accessed August 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11467015/
  2. Vorasidenib, a Dual Inhibitor of Mutant IDH1/2, in Recurrent or ..., accessed August 28, 2025, https://aacrjournals.org/clincancerres/article/27/16/4491/671595/Vorasidenib-a-Dual-Inhibitor-of-Mutant-IDH1-2-in
  3. INDIGO: A global, randomized, double-blinded, phase 3 study of vorasidenib versus placebo in patients with residual or recurrent grade 2 glioma with an IDH1/2 mutation. | Journal of Clinical Oncology - ASCO Publications, accessed August 28, 2025, https://ascopubs.org/doi/10.1200/JCO.2023.41.17_suppl.LBA1
  4. Characteristics and Outcomes of Patients With IDH-Mutant Grade 2 ..., accessed August 28, 2025, https://www.neurology.org/doi/10.1212/WNL.0000000000213797
  5. Fundamental Research Paved the Way for the Development of Vorasidenib, a New Clinical Treatment for IDH-Mutant Low-Grade Gliomas - National Cancer Institute, accessed August 28, 2025, https://www.cancer.gov/about-nci/organization/dcb/progress/cancer-biology-and-vorasidenib-development
  6. Glioma Study - End Brain Cancer Initiative, accessed August 28, 2025, https://endbraincancer.org/glioma-study/
  7. Vorasidenib in IDH1/2-mutant low-grade glioma: the grey zone of patient's selection - PMC, accessed August 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10809174/
  8. Vorasidenib - Wikipedia, accessed August 28, 2025, https://en.wikipedia.org/wiki/Vorasidenib
  9. Glioma Mechanism of Disease & VORANIGO® (vorasidenib) MOA, accessed August 28, 2025, https://www.voranigohcp.com/about/mod-moa
  10. Vorasidenib | C14H13ClF6N6 | CID 117817422 - PubChem, accessed August 28, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Vorasidenib
  11. Vorasidenib: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed August 28, 2025, https://go.drugbank.com/drugs/DB17097
  12. Management for Different Glioma Subtypes: Are All Low-Grade Gliomas Created Equal?, accessed August 28, 2025, https://ascopubs.org/doi/10.1200/EDBK_238353
  13. FDA approves new therapy for glioma patients for first time in decades, accessed August 28, 2025, https://www.dana-farber.org/newsroom/news-releases/2024/fda-approves-new-therapy-for-glioma-patients-for-first-time-in-decades
  14. Vorasidenib (AG-881): A First-in-Class, Brain-Penetrant Dual Inhibitor of Mutant IDH1 and 2 for Treatment of Glioma - PMC - PubMed Central, accessed August 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7025383/
  15. Vorasidenib (AG-881): A First-in-Class, Brain-Penetrant Dual Inhibitor of Mutant IDH1 and 2 for Treatment of Glioma | ACS Medicinal Chemistry Letters - ACS Publications, accessed August 28, 2025, https://pubs.acs.org/doi/10.1021/acsmedchemlett.9b00509
  16. Vorasidenib (AG881) - cas 1644545-52-7 - DC Chemicals, accessed August 28, 2025, https://www.dcchemicals.com/product_show-AG881.html
  17. Voranigo (vorasidenib) FDA Approval History - Drugs.com, accessed August 28, 2025, https://www.drugs.com/history/voranigo.html
  18. Compound: VORASIDENIB (CHEMBL4279047) - ChEMBL - EMBL-EBI, accessed August 28, 2025, https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL4279047
  19. Vorasidenib | AG-881 | CAS#1644545-52-7 | IDH1 Inhibitor - MedKoo Biosciences, accessed August 28, 2025, https://www.medkoo.com/products/15117
  20. vorasidenib | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed August 28, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=10663
  21. Vorasidenib (AG-881) | mIDH1/2 Inhibitor - Ambeed.com, accessed August 28, 2025, https://www.ambeed.com/products/vorasidenib.html
  22. Vorasidenib (AG-881, CAS Number: 1644545-52-7) - Cayman Chemical, accessed August 28, 2025, https://www.caymanchem.com/product/28462/vorasidenib
  23. www.cancer.gov, accessed August 28, 2025, https://www.cancer.gov/about-nci/organization/dcb/progress/cancer-biology-and-vorasidenib-development#:~:text=In%20a%20surgical%20study%20investigating,with%20the%20mutant%20IDH1%20enzyme.
  24. VORANIGO® (vorasidenib) tablets, for oral use - accessdata.fda.gov, accessed August 28, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/218784s000lbl.pdf
  25. Dosing & Administration - VORANIGO® (vorasidenib), accessed August 28, 2025, https://www.voranigohcp.com/dosing-management
  26. Vorasidenib, a Dual Inhibitor of Mutant IDH1/2, in Recurrent or Progressive Glioma; Results of a First-in-Human Phase I Trial - PubMed, accessed August 28, 2025, https://pubmed.ncbi.nlm.nih.gov/34078652/
  27. FDA approves vorasidenib for Grade 2 astrocytoma or ..., accessed August 28, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-vorasidenib-grade-2-astrocytoma-or-oligodendroglioma-susceptible-idh1-or-idh2-mutation
  28. FDA and EMA Accept Vorasidenib Regulatory Submissions for the Treatment of IDH-mutant diffuse glioma - PR Newswire, accessed August 28, 2025, https://www.prnewswire.com/news-releases/fda-and-ema-accept-vorasidenib-regulatory-submissions-for-the-treatment-of-idh-mutant-diffuse-glioma-302064504.html
  29. Study Details | Vorasidenib Maintenance for IDH Mutant Astrocytoma - ClinicalTrials.gov, accessed August 28, 2025, https://www.clinicaltrials.gov/study/NCT06809322
  30. Servier Receives Regulatory Filing Acceptances from FDA and EMA for Vorasidenib in the Treatment of IDH-Mutant Diffuse Glioma | Blog, accessed August 28, 2025, https://servier.us/blog/vorasidenib-ema-fda-release/
  31. Vorasidenib: First Approval - PMC - PubMed Central, accessed August 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11628568/
  32. Servier's VORANIGO® (vorasidenib) tablets receives FDA approval as first targeted therapy for Grade 2 IDH-mutant glioma | Blog, accessed August 28, 2025, https://servier.us/blog/serviers-voranigo-vorasidenib-tablets-receives-fda-approval-as-first-targeted-therapy-for-grade-2-idh-mutant-glioma/
  33. Vorasidenib for IDH-mutant grade 2 gliomas: clinical advances and future directions, accessed August 28, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2025.1628195/full
  34. Voranigo | European Medicines Agency (EMA), accessed August 28, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/voranigo
  35. Voranigo; INN-vorasidenib - European Medicines Agency, accessed August 28, 2025, https://www.ema.europa.eu/en/documents/smop-initial/chmp-summary-positive-opinion-voranigo_en.pdf
  36. CHMP Recommends EU Approval of Vorasidenib for IDH1/2-Mutant Grade 2 Glioma, accessed August 28, 2025, https://www.onclive.com/view/chmp-recommends-eu-approval-of-vorasidenib-for-idh1-2-mutant-grade-2-glioma
  37. VORANIGO (vorasidenib), FDA Approved for the Treatment of Patients with IDH-mutant Diffuse Glioma, Available at Biologics by McKesson, accessed August 28, 2025, https://www.mckesson.com/about-us/newsroom/press-releases/2024/voranigo-vorasidenib-fda-approved-for-the-treatment-of-patients-with-idh-mutant-diffuse-glioma-available-at-biologics-by-mckesson/
  38. Clinical Trials Using Vorasidenib - NCI, accessed August 28, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/vorasidenib?pn=1
  39. Study of Vorasidenib and Pembrolizumab Combination in Recurrent or Progressive Enhancing IDH-1 Mutant Glioma | UCSF Brain Tumor Center, accessed August 28, 2025, https://braintumorcenter.ucsf.edu/clinical-trial/study-vorasidenib-and-pembrolizumab-combination-recurrent-or-progressive-enhancing
  40. Study of Vorasidenib and Pembrolizumab Combination in Recurrent or Progressive IDH-1 Mutant Glioma - Mayo Clinic, accessed August 28, 2025, https://www.mayo.edu/research/clinical-trials/cls-20576295

Published at: August 28, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.