C18H15ClN4O2
1887014-12-1
Refractory Acute Myeloid Leukemia (AML), Relapsed Acute Myelogenous Leukemia (AML)
Olutasidenib, marketed under the brand name Rezlidhia, is an orally administered, potent, and selective small-molecule inhibitor of mutated isocitrate dehydrogenase-1 (mIDH1).[1] It represents a significant advancement in the targeted therapy of acute myeloid leukemia (AML). The drug's mechanism of action is precisely targeted, inhibiting the neomorphic enzymatic activity of mIDH1, which is responsible for the oncogenic production of 2-hydroxyglutarate (2-HG). By suppressing 2-HG, Olutasidenib alleviates the epigenetic blockade characteristic of mIDH1-driven malignancies, thereby restoring normal hematopoietic cell differentiation.[4]
The clinical efficacy of Olutasidenib was definitively established in the pivotal cohort of the Phase 2 study 2102-HEM-101, which evaluated its use in adult patients with relapsed or refractory (R/R) AML harboring a susceptible IDH1 mutation. The trial met its primary endpoint, demonstrating a composite complete remission (CR) and CR with partial hematologic recovery (CRh) rate of 35%. The most striking feature of this response was its unprecedented durability, with a median duration of 25.9 months—a substantial improvement over existing therapies in this patient population.[6] This sustained effect suggests a profound and lasting impact on the underlying disease biology, potentially altering the natural history of the disease for responding patients.
This significant clinical benefit is balanced by a distinct and serious safety profile that requires diligent clinical management. The prescribing information for Olutasidenib carries a boxed warning for Differentiation Syndrome (DS), a potentially fatal complication arising from the rapid proliferation and differentiation of myeloid cells, which is mechanistically linked to the drug's therapeutic effect. Additionally, hepatotoxicity is a prominent risk, necessitating a rigorous schedule of liver function monitoring and clear protocols for dose modification or discontinuation.[1]
Based on the robust efficacy data, Olutasidenib was granted approval by the U.S. Food and Drug Administration (FDA) on December 1, 2022, for the treatment of adult patients with R/R AML with a susceptible IDH1 mutation, as detected by an FDA-approved companion diagnostic test.[1] This approval positions Olutasidenib as a critical therapeutic option for a molecularly defined subset of AML patients with historically poor prognoses, offering the potential for long-term, durable remissions that were previously unattainable. Its distinct efficacy and safety profile, particularly when compared to the other approved IDH1 inhibitor, Ivosidenib, provides clinicians with a new tool that allows for more nuanced, patient-specific treatment decisions based on comorbidities and therapeutic goals.
This section provides a definitive summary of the nomenclature, identifiers, and physicochemical characteristics of Olutasidenib, establishing a foundational reference for the molecule.
Olutasidenib is identified through a variety of systematic names and database codes, ensuring its unambiguous recognition across scientific, clinical, and regulatory domains.
Olutasidenib is classified as a small molecule drug, characterized by the following properties [18]:
A consolidated list of key identifiers is provided in Table 2.1 for ease of reference. This table serves as a comprehensive cross-reference, linking the various names and codes used in different contexts, from chemical registries to pharmacological databases.
Table 2.1: Summary of Physicochemical Properties and Identifiers
| Property/Identifier | Value | Source(s) | 
|---|---|---|
| Generic Name | Olutasidenib | 1 | 
| Brand Name | Rezlidhia | 2 | 
| CAS Number | 1887014-12-1 | 1 | 
| DrugBank ID | DB16267 | 1 | 
| PubChem CID | 118955396 | 2 | 
| UNII | 0T4IMT8S5Z | 1 | 
| Chemical Formula | $C_{18}H_{15}ClN_{4}O_{2}$ | 2 | 
| Molar Mass | 354.79 g·mol⁻¹ | 2 | 
| IUPAC Name | 5-{amino}-1-methyl-6-oxo-1H-pyridine-2-carbonitrile | 2 | 
| SMILES String | C[C@H](NC1=CC=C(C#N)N(C)C1=O)C1=CC2=C(NC1=O)C=CC(Cl)=C2 | 19 | 
| InChI Key | NEQYWYXGTJDAKR-JTQLQIEISA-N | 17 | 
The therapeutic activity of Olutasidenib is rooted in its ability to precisely target a specific metabolic vulnerability created by mutations in the isocitrate dehydrogenase 1 (IDH1) gene. Its mechanism is not cytotoxic in the manner of traditional chemotherapy but rather cytodifferentiative, aiming to reverse the oncogenic state and restore normal cellular function.
Under normal physiological conditions, the IDH family of enzymes plays a pivotal role in cellular metabolism. The IDH1 isoform is located in the cytoplasm and peroxisomes, while IDH2 and IDH3 reside in the mitochondria. These enzymes are central to the tricarboxylic acid (TCA) or Krebs cycle, where they catalyze the oxidative decarboxylation of isocitrate to produce α-ketoglutarate (α-KG) and NADPH.[5] The product, α-KG, is a critical metabolic intermediate that serves as a substrate for a large family of dioxygenase enzymes involved in diverse cellular processes, including epigenetic regulation.[7]
In certain malignancies, including AML, gliomas, and cholangiocarcinoma, somatic point mutations arise in the IDH1 gene. These mutations occur in approximately 6-16% of AML cases and are most frequently found at the arginine 132 (R132) residue of the enzyme's active site.[1] These are not loss-of-function mutations; instead, they are gain-of-function mutations that confer a novel, or neomorphic, enzymatic activity.[5] While losing its ability to produce α-KG from isocitrate, the mutated enzyme gains the capacity to catalyze the NADPH-dependent reduction of α-KG into the oncometabolite (R)-2-hydroxyglutarate (2-HG).[4]
The massive accumulation of 2-HG in cancer cells is the central driver of mIDH1-mediated oncogenesis. Due to its structural similarity to α-KG, 2-HG acts as a competitive inhibitor of numerous α-KG-dependent dioxygenases.[5] Key targets of this inhibition include the TET (ten-eleven translocation) family of DNA demethylases and Jumonji-domain-containing histone demethylases.[5] The resulting enzymatic inhibition leads to widespread epigenetic dysregulation, characterized by DNA and histone hypermethylation. This altered epigenetic landscape silences genes required for normal cellular maturation, effectively blocking hematopoietic differentiation and locking progenitor cells in an immature, proliferative state that ultimately leads to the development of leukemia.[1]
Olutasidenib was specifically designed as a potent and highly selective oral inhibitor of the mutated IDH1 enzyme.[1] Unlike competitive inhibitors that would bind to the active site, Olutasidenib is a non-competitive, allosteric inhibitor. It binds to a distinct hydrophobic pocket located near the interface of the IDH1 homodimer.[22] This binding stabilizes the enzyme in an open, inactive conformation, which prevents the conformational change necessary for its neomorphic catalytic activity and subsequent production of 2-HG.[22]
This allosteric mechanism contributes to the drug's remarkable selectivity. In vitro studies have confirmed that Olutasidenib potently inhibits a range of clinically relevant IDH1 R132 mutations—including R132H, R132C, R132G, R132L, and R132S—while showing negligible activity against wild-type IDH1 or mutated IDH2 enzymes.[6] This high degree of selectivity minimizes off-target effects on normal cellular metabolism, which relies on the proper functioning of wild-type IDH enzymes. The specific inhibitory activity is quantified in Table 3.1.
Table 3.1: In Vitro Inhibitory Activity ($IC_{50}$) of Olutasidenib Against mIDH1 Variants and Selectivity Profile
| Target Enzyme | IC50 Value | Source(s) | 
|---|---|---|
| IDH1-R132H | 21.1 - 21.2 nM (0.021 µM) | 17 | 
| IDH1-R132C | 114 nM (0.114 µM) | 17 | 
| Wild-Type IDH1 | > 20 µM (22.4 µM) | 17 | 
| IDH2-R172K | 27.3 µM | 17 | 
| IDH2-R140Q | > 100 µM | 17 | 
By potently inhibiting mIDH1, Olutasidenib leads to a rapid and sustained reduction in intracellular and plasma levels of 2-HG.[6] In clinical studies, patients treated with the approved dose experienced a mean reduction of 59.1% in plasma 2-HG.[6] The depletion of this oncometabolite removes the competitive inhibition of α-KG-dependent dioxygenases. This allows for the reactivation of enzymes like TET2, leading to the reversal of the aberrant hypermethylation state and the restoration of normal gene expression patterns.[5]
The ultimate clinical consequence of this epigenetic reprogramming is the induction of cellular differentiation. The blockade on maturation is lifted, and leukemic blasts are able to differentiate into mature, functional myeloid cells.[3] This process is reflected clinically by a reduction in bone marrow blast counts, an improvement in peripheral blood counts, and the achievement of clinical remission.
The cytodifferentiative mechanism, as opposed to a cytotoxic one, has direct and profound clinical implications. First, it explains the observed timeline to clinical response. The process of reversing epigenetic changes and allowing for the maturation of a new wave of healthy myeloid cells is not instantaneous, which accounts for the median time to response of 1.9 months.[3] This understanding is critical for clinicians to manage patient expectations and avoid premature discontinuation of a therapy that requires time to manifest its full effect. Second, this mechanism is directly linked to the drug's most significant on-target toxicity. The intended therapeutic effect—the rapid proliferation and differentiation of myeloid cells—can become pathologically exaggerated, leading to a systemic inflammatory response known as Differentiation Syndrome.[3] This direct connection between the drug's therapeutic action and its primary toxicity underscores the importance of vigilant monitoring for this specific adverse event.
The clinical utility of Olutasidenib is governed by its pharmacokinetic profile, which describes its absorption, distribution, metabolism, and excretion (ADME), and its pharmacodynamic effects, which relate drug concentration to its biological activity.
Olutasidenib is an orally bioavailable agent.[1] Following oral administration of a single 150 mg dose, the median time to reach maximum plasma concentration ($T_{max}$) is approximately 4 hours.[6] The absorption of Olutasidenib is significantly influenced by food. Administration with a high-fat, high-calorie meal results in a dramatic increase in drug exposure, with the mean maximum concentration ($C_{max}$) increasing by 191% and the total exposure (Area Under the Curve, AUC) increasing by 83%.[6] This substantial food effect is a critical factor in clinical practice, as uncontrolled variations in exposure could lead to increased toxicity. This finding underpins the strict administration instruction to take the drug on an empty stomach to ensure consistent and predictable absorption.[11]
Once absorbed, Olutasidenib distributes widely throughout the body, as indicated by a large apparent volume of distribution ($V_{d}$) of 319 L.[26] It is highly bound to plasma proteins, with a binding fraction of approximately 93%.[26] A key feature of Olutasidenib's distribution is its ability to penetrate the blood-brain barrier (BBB).[1] This characteristic is of significant clinical interest, as it makes Olutasidenib a viable candidate for treating mIDH1-driven central nervous system malignancies, such as gliomas, and is a focus of ongoing clinical investigation.[1]
Olutasidenib undergoes extensive metabolism in the liver. The primary metabolic pathway, accounting for approximately 90% of its clearance, is mediated by the Cytochrome P450 3A4 (CYP3A4) enzyme.[3] Minor metabolic contributions are made by other CYP isoforms, including CYP2C8, CYP2C9, CYP1A2, and CYP2C19.[14] The metabolic processes include N-dealkylation, demethylation, and oxidation, followed by glucuronidation.[26] This heavy reliance on CYP3A4 for metabolism makes Olutasidenib susceptible to significant drug-drug interactions.
The elimination of Olutasidenib and its metabolites occurs primarily through the fecal route. Approximately 75% of an administered dose is recovered in the feces, with 35% of that being the unchanged parent drug.[14] A smaller portion, about 17%, is excreted in the urine, with only 1% as unchanged drug.[14] The drug has a long terminal elimination half-life ($t_{1/2}$) of approximately 67 hours.[3] This long half-life, combined with twice-daily dosing, leads to significant drug accumulation, with observed accumulation ratios between 7.7 and 9.5. Steady-state plasma concentrations are typically reached within 14 days of initiating therapy.[14] The slow clearance has important implications for managing adverse events; upon drug discontinuation, washout will be gradual, reinforcing the need for proactive and primary interventions (e.g., corticosteroids for DS) rather than relying solely on holding the drug for immediate effect.
The pharmacodynamic effect of Olutasidenib is directly linked to its mechanism of action. The recommended dose of 150 mg twice daily has been shown to achieve maximal suppression of plasma 2-HG levels, with a mean reduction of 59.1% from baseline, confirming on-target biological activity.[6] Exposure-response analyses have revealed a positive correlation between higher plasma concentrations of Olutasidenib and an increased probability of experiencing Grade 3 or higher Differentiation Syndrome and hepatotoxicity.[6] This relationship between exposure and toxicity further highlights the clinical importance of the significant food effect; taking Olutasidenib with food could inadvertently shift a patient from a therapeutic exposure range into a toxic one, making the instruction to dose on an empty stomach a critical safety measure.
Table 4.1: Key Pharmacokinetic Parameters of Olutasidenib
| Parameter | Value | Source(s) | 
|---|---|---|
| Time to Peak ($T_{max}$) | Median ~4 hours | 6 | 
| Steady-State $C_{max}$ | 3,573 ng/mL (45.6% CV) | 14 | 
| Steady-State Daily AUC | 43,050 ng·h/mL (34.0% CV) | 14 | 
| Terminal Half-Life ($t_{1/2}$) | ~67 hours (51.2% CV) | 6 | 
| Apparent Volume of Distribution ($V_{d}$) | 319 L | 26 | 
| Apparent Oral Clearance (CL/F) | 4 L/h (60.5% CV) | 14 | 
| Plasma Protein Binding | ~93% | 26 | 
| Primary Metabolism | CYP3A4 (~90%) | 14 | 
| Primary Excretion Route | Feces (~75%) | 14 | 
The approval of Olutasidenib was based on the compelling efficacy demonstrated in a pivotal cohort of patients with relapsed or refractory (R/R) AML with a susceptible IDH1 mutation. The data from this study established the drug's ability to induce deep and exceptionally durable remissions in a population with a historically poor prognosis.
The clinical evidence for Olutasidenib's efficacy is derived from Study 2102-HEM-101, a Phase 1/2, open-label, single-arm, multicenter clinical trial.[3] The registrational data came from a specific cohort within this study comprising 153 adult patients with R/R AML. Of these, 147 were evaluable for the efficacy analysis.[6]
Key eligibility criteria included a confirmed susceptible IDH1 mutation, detected centrally using the Abbott RealTime™ IDH1 Assay, which was concurrently approved as a companion diagnostic.[8] The patient population was characteristic of R/R AML, with a median age of 71 years and having been heavily pre-treated with a median of two prior therapeutic regimens.[9] Patients in this cohort received Olutasidenib monotherapy at the recommended Phase 2 dose of 150 mg orally twice daily.[6]
The study successfully met its primary endpoints, demonstrating both a high rate of response and remarkable durability.
The clinical benefit of Olutasidenib extended beyond the primary endpoint to other important measures of efficacy.
Further analyses and related studies have provided additional context for Olutasidenib's clinical activity.
Table 5.1: Efficacy Results from the Pivotal Cohort of Study 2102-HEM-101
| Endpoint | Value | Source(s) | 
|---|---|---|
| CR+CRh Rate (95% CI) | 35% (27-43%) | 6 | 
| CR Rate | 32% | 8 | 
| CRh Rate | 2.7% | 8 | 
| Median Time to CR+CRh | 1.9 months | 3 | 
| Median Duration of CR+CRh (95% CI) | 25.9 months (13.5-NR) | 6 | 
| Overall Response Rate (ORR) | 48% | 9 | 
| Median Overall Survival (OS) | 11.6 months | 3 | 
| Rate of Transfusion Independence | 34% (in baseline dependent patients) | 6 | 
While Olutasidenib offers significant efficacy, its use is associated with a distinct set of serious adverse events that require a high degree of clinical vigilance, proactive monitoring, and specific management strategies. The safety profile is manageable but necessitates a thorough understanding of the risks by the prescribing clinician.
The most critical safety concern with Olutasidenib is Differentiation Syndrome, a risk highlighted by a boxed warning in the prescribing information.[8]
Hepatotoxicity is another significant warning and precaution associated with Olutasidenib therapy.
Beyond DS and hepatotoxicity, the overall safety profile of Olutasidenib is characterized by a range of common adverse events and hematologic toxicities.
The safety profile necessitates that Olutasidenib be prescribed by specialists in settings equipped for intensive monitoring. The decision to use this drug must be coupled with a commitment to the required surveillance protocols to ensure patient safety. This is particularly relevant when considering the choice between Olutasidenib and the other approved IDH1 inhibitor, Ivosidenib. While their efficacy rates are similar, their key non-hematologic toxicities differ: hepatotoxicity for Olutasidenib versus QTc interval prolongation for Ivosidenib.[3] This distinction creates a clear framework for clinical decision-making. For a patient with significant pre-existing liver disease, Ivosidenib might be the preferred agent. Conversely, for a patient with a history of cardiac arrhythmias or who is taking other QTc-prolonging medications, Olutasidenib would represent the safer choice from a cardiovascular perspective. This allows for treatment selection to be tailored to the individual patient's comorbidity profile.
Table 6.1: Incidence of Common Adverse Reactions (≥20%) in Study 2102-HEM-101
| Adverse Reaction | All Grades (%) | Grade 3-4 (%) | Source(s) | 
|---|---|---|---|
| Nausea | 38% | 0% | 8 | 
| Fatigue/Malaise | 36% | 3% | 8 | 
| Arthralgia | 28% | 3% | 8 | 
| Constipation | 26% | 0% | 8 | 
| Leukocytosis | 25% | 9% | 8 | 
| Dyspnea | 24% | 5% | 8 | 
| Fever (Pyrexia) | 24% | 1% | 8 | 
| Rash | 24% | 1% | 8 | 
| Mucositis | 23% | 3% | 8 | 
| Diarrhea | 20% | 1% | 8 | 
| Transaminitis | 20% | 12% | 8 | 
Table 6.2: Guidelines for Dosage Modification in Response to Key Toxicities
| Toxicity | Recommended Action | Source(s) | 
|---|---|---|
| Differentiation Syndrome | Withhold Olutasidenib. Initiate systemic corticosteroids (e.g., dexamethasone 10 mg IV q12h) and hemodynamic monitoring. Resume at 150 mg BID after resolution. | 10 | 
| Hepatotoxicity (Grade 3) | Withhold Olutasidenib and monitor LFTs. Upon resolution to baseline or Grade 1, resume at a reduced dose of 150 mg once daily. May re-escalate to 150 mg BID if stable for ≥28 days. | 10 | 
| Hepatotoxicity (Grade 4) | Permanently discontinue Olutasidenib. | 10 | 
| Other Grade ≥3 Adverse Effects | Interrupt dosing until toxicity resolves to Grade ≤2. Resume at 150 mg once daily. May re-escalate to 150 mg BID if stable for ≥1 week. Discontinue if recurs at 150 mg once daily. | 10 | 
The safe and effective use of Olutasidenib requires strict adherence to specific guidelines regarding its dosage, administration, and the management of potential drug-drug interactions.
Patient education on proper administration is crucial to ensure consistent drug exposure and minimize risks.
Olutasidenib's metabolism and its own effect on metabolic enzymes create a complex profile of potential drug-drug interactions that must be carefully managed.
The dual role of Olutasidenib as both a major substrate and a moderate inducer of the critical CYP3A4 enzyme pathway necessitates a meticulous and ongoing review of all concomitant medications for any patient receiving the drug. This complex interaction profile underscores the vital role of clinical pharmacists as part of the multidisciplinary care team. They can provide essential expertise in identifying potential interactions, recommending alternative medications, and advising on necessary dose adjustments to ensure both the efficacy of Olutasidenib and the safety and effectiveness of the patient's other medications.
The regulatory journey of Olutasidenib reflects its targeted nature and its role in addressing an unmet need in a rare disease. Its approval in the United States has established its place in the treatment armamentarium, while its development program continues to explore its potential in other settings.
No approvals for Olutasidenib have been reported from other major regulatory bodies such as the Therapeutic Goods Administration (TGA) in Australia, Health Canada, or the Spanish Agency of Medicines and Medical Devices (CIMA AEMPS).[34]
The clinical development of Olutasidenib is actively pursuing a "pipeline-in-a-pill" strategy, aiming to expand its utility based on its molecular target rather than being confined to a single disease histology.
To fully appreciate the clinical value of Olutasidenib, it is essential to place it within the current therapeutic landscape for mIDH1-mutated R/R AML. This involves a direct comparison with the other approved agent in its class, Ivosidenib, and an analysis of its role in the overall treatment algorithm.
In the absence of a head-to-head randomized controlled trial, any comparison between Olutasidenib and Ivosidenib must be made by interpreting data from their respective single-arm pivotal studies. While such cross-trial comparisons should be approached with caution due to potential differences in patient populations and study conduct, they can reveal important distinctions in the drugs' clinical profiles.[3]
This comparative analysis reveals a nuanced clinical choice. A clinician is presented with two agents with similar response rates but a stark difference in response durability and primary toxicities. This suggests a "high-risk, high-reward" dynamic. The potential reward with Olutasidenib is a significantly longer and more durable remission, which could fundamentally alter the disease course. However, this reward is balanced by the risk of managing potentially severe hepatotoxicity, which requires a more intensive monitoring schedule. The choice between the two agents is therefore likely to be driven by a careful assessment of the individual patient's goals of care, comorbidities, and the clinical team's capacity for vigilant monitoring.
Olutasidenib is recognized as a standard of care for its approved indication. It is included in the National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines as a category 2A recommended targeted therapy for adult patients with R/R AML with an IDH1 mutation.[3] It provides a critical and effective option for patients who have failed prior lines of therapy, including both intensive chemotherapy and the now-commonplace venetoclax-based regimens.[7] The demonstrated efficacy in the post-venetoclax setting is particularly important, as it establishes a clear and evidence-based sequencing pathway for this challenging patient population.
Table 9.1: Comparative Overview of Olutasidenib and Ivosidenib for R/R mIDH1 AML
| Feature | Olutasidenib (Rezlidhia) | Ivosidenib (Tibsovo) | 
|---|---|---|
| CR+CRh Rate | 35% | 32.8% | 
| Median Duration of CR+CRh | 25.9 months | 8.2 months | 
| Median Overall Survival | 11.6 months | 8.8 months | 
| Key Non-Hematologic Toxicity | Hepatotoxicity | QTc Prolongation | 
| Dosing Schedule | 150 mg twice daily (empty stomach) | 500 mg once daily (with or without food) | 
| Source(s) | 3 | 3 | 
Olutasidenib (Rezlidhia) represents a significant and welcome addition to the therapeutic armamentarium for acute myeloid leukemia. Its development and approval have solidified the role of targeted metabolic inhibition as a key strategy in precision oncology.
The clinical value of Olutasidenib is defined by a compelling risk-benefit profile. It is a highly effective targeted therapy for adult patients with relapsed or refractory AML with a susceptible IDH1 mutation, offering the potential for deep and, most notably, exceptionally durable remissions. The median duration of response of over two years in this poor-prognosis population is a transformative achievement. This profound benefit, however, is balanced by a safety profile that demands careful management. The boxed warning for Differentiation Syndrome and the significant risk of hepatotoxicity necessitate vigilant patient monitoring and a clear, proactive strategy for managing these potentially life-threatening adverse events. When used appropriately by experienced clinicians in a suitable setting, the benefits of Olutasidenib in inducing long-term, durable disease control are substantial.
The successful development of Olutasidenib as a monotherapy has paved the way for numerous avenues of future research aimed at optimizing its use and expanding its applications.
In conclusion, Olutasidenib is a paradigm of modern, molecularly targeted cancer therapy. Its approval has provided a new standard of care for a specific subset of AML patients, offering a chance for long-term disease control that was previously out of reach. Beyond its direct clinical impact, the story of Olutasidenib has deepened the scientific community's understanding of the intricate links between cellular metabolism, epigenetics, and oncogenesis. The future of Olutasidenib will be defined by research focused on intelligent combination strategies, optimal sequencing, and the expansion of its use to the full spectrum of mIDH1-mutated malignancies.
Published at: October 24, 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.