MedPath

Olutasidenib Advanced Drug Monograph

Published:Oct 24, 2025

Generic Name

Olutasidenib

Brand Names

Rezlidhia

Drug Type

Small Molecule

Chemical Formula

C18H15ClN4O2

CAS Number

1887014-12-1

Associated Conditions

Refractory Acute Myeloid Leukemia (AML), Relapsed Acute Myelogenous Leukemia (AML)

Olutasidenib (Rezlidhia): A Comprehensive Monograph on the Mutant IDH1 Inhibitor for Relapsed/Refractory Acute Myeloid Leukemia

Executive Summary

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.

Drug Profile and Chemical Properties

This section provides a definitive summary of the nomenclature, identifiers, and physicochemical characteristics of Olutasidenib, establishing a foundational reference for the molecule.

Nomenclature and Identifiers

Olutasidenib is identified through a variety of systematic names and database codes, ensuring its unambiguous recognition across scientific, clinical, and regulatory domains.

  • Generic Name: The internationally recognized nonproprietary name (INN) for the compound is Olutasidenib.[1]
  • Brand Name: In the United States, Olutasidenib is marketed by Rigel Pharmaceuticals under the trade name Rezlidhia®.[2]
  • Development Codes: During its preclinical and clinical development, the compound was referred to as FT-2102 or FT 2102.[1]
  • Systematic (IUPAC) Name: The formal chemical name according to the International Union of Pure and Applied Chemistry (IUPAC) nomenclature is 5-{amino}-1-methyl-6-oxo-1H-pyridine-2-carbonitrile.[2]

Chemical and Physical Data

Olutasidenib is classified as a small molecule drug, characterized by the following properties [18]:

  • Chemical Formula: The molecular formula is $C_{18}H_{15}ClN_{4}O_{2}$.[2]
  • Molar Mass: The molar mass (or formula weight) is 354.79 g·mol⁻¹.[2]
  • Physical Form: In its purified state, Olutasidenib exists as a crystalline solid.[17]
  • Solubility: The compound is soluble in dimethyl sulfoxide (DMSO), a common solvent used for in vitro biological assays.[17]
  • Structural Identifiers: For computational and database purposes, the molecule is represented by standardized linear notations:
  • SMILES (Simplified Molecular-Input Line-Entry System): C[C@H](NC1=CC=C(C#N)N(C)C1=O)C1=CC2=C(NC1=O)C=CC(Cl)=C2.[19]
  • InChI Key (International Chemical Identifier Key): NEQYWYXGTJDAKR-JTQLQIEISA-N.[17]

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/IdentifierValueSource(s)
Generic NameOlutasidenib1
Brand NameRezlidhia2
CAS Number1887014-12-11
DrugBank IDDB162671
PubChem CID1189553962
UNII0T4IMT8S5Z1
Chemical Formula$C_{18}H_{15}ClN_{4}O_{2}$2
Molar Mass354.79 g·mol⁻¹2
IUPAC Name5-{amino}-1-methyl-6-oxo-1H-pyridine-2-carbonitrile2
SMILES StringC[C@H](NC1=CC=C(C#N)N(C)C1=O)C1=CC2=C(NC1=O)C=CC(Cl)=C219
InChI KeyNEQYWYXGTJDAKR-JTQLQIEISA-N17

Mechanism of Action: Targeting the IDH1 Mutation in Oncogenesis

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.

The Role of Isocitrate Dehydrogenase (IDH) in Normal Cellular Metabolism

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]

Oncogenic Transformation via Mutant IDH1 (mIDH1)

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 Pathophysiological Role of 2-HG

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 as a Selective, Allosteric Inhibitor of mIDH1

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 EnzymeIC50​ ValueSource(s)
IDH1-R132H21.1 - 21.2 nM (0.021 µM)17
IDH1-R132C114 nM (0.114 µM)17
Wild-Type IDH1> 20 µM (22.4 µM)17
IDH2-R172K27.3 µM17
IDH2-R140Q> 100 µM17

Downstream Effects and Restoration of Normal Hematopoiesis

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.

Pharmacokinetics and Pharmacodynamics

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.

Absorption

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]

Distribution

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]

Metabolism

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.

Excretion

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.

Pharmacodynamics

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

ParameterValueSource(s)
Time to Peak ($T_{max}$)Median ~4 hours6
Steady-State $C_{max}$3,573 ng/mL (45.6% CV)14
Steady-State Daily AUC43,050 ng·h/mL (34.0% CV)14
Terminal Half-Life ($t_{1/2}$)~67 hours (51.2% CV)6
Apparent Volume of Distribution ($V_{d}$)319 L26
Apparent Oral Clearance (CL/F)4 L/h (60.5% CV)14
Plasma Protein Binding~93%26
Primary MetabolismCYP3A4 (~90%)14
Primary Excretion RouteFeces (~75%)14

Clinical Efficacy in Relapsed/Refractory Acute Myeloid Leukemia

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.

Pivotal Trial Design: Study 2102-HEM-101 (NCT02719574)

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]

Primary Efficacy Endpoints

The study successfully met its primary endpoints, demonstrating both a high rate of response and remarkable durability.

  • CR+CRh Rate: The primary efficacy endpoint was the rate of composite complete remission (CR) plus CR with partial hematologic recovery (CRh). The observed CR+CRh rate was 35% (95% Confidence Interval [CI]: 27-43%).[6] This composite rate was driven primarily by deep remissions, with a 32% rate of CR and a 2.7% rate of CRh.[8]
  • Duration of CR+CRh: The median duration of CR+CRh was 25.9 months (95% CI: 13.5-not reached).[6] This finding is particularly significant, as a duration of response exceeding two years is exceptional in the R/R AML setting, where survival is often measured in months. For the subset of patients who achieved a full CR, the median duration of response was even longer at 28.1 months.[27]
  • Time to Response: The therapeutic effect was observed relatively early, with a median time to first CR or CRh of 1.9 months.[3]

Secondary and Other Efficacy Endpoints

The clinical benefit of Olutasidenib extended beyond the primary endpoint to other important measures of efficacy.

  • Overall Response Rate (ORR): The ORR, a broader measure of anti-leukemic activity that includes partial remissions and morphologic leukemia-free states, was 48%.[9]
  • Overall Survival (OS): In this heavily pre-treated population, the median overall survival was 11.6 months.[3]
  • Transfusion Independence: Olutasidenib demonstrated a clinically meaningful impact on patients' quality of life by reducing the burden of blood transfusions. Of the 86 patients who were dependent on red blood cell (RBC) and/or platelet transfusions at baseline, 29 (34%) achieved transfusion independence for a period of at least 56 consecutive days.[6] This benefit was not limited to patients achieving deep remissions; it was observed across all response groups, indicating that even patients with lesser responses could experience this tangible improvement.[9] This broadens the definition of clinical benefit beyond formal remission criteria to include patient-centric outcomes.

Subgroup Analyses and Other Relevant Studies

Further analyses and related studies have provided additional context for Olutasidenib's clinical activity.

  • Prior Venetoclax Exposure: A critical question in modern AML therapy is how to treat patients who have relapsed after treatment with venetoclax-based regimens. The trial data showed that Olutasidenib's efficacy was maintained in this difficult-to-treat population, with a CR/CRh rate of 33%.[3] This finding provides clinicians with a clear, evidence-based sequencing option for a growing area of unmet need.
  • Combination Therapy: The Phase 1 portion of the study explored Olutasidenib in combination with the hypomethylating agent azacitidine. In treatment-naïve AML patients unfit for intensive chemotherapy, this combination yielded a high overall response rate of 77%.[32]
  • Maintenance Therapy: Given its favorable oral administration and durability of response, Olutasidenib is being explored as a maintenance therapy. Preliminary data from a small cohort of patients who received Olutasidenib after achieving remission with induction therapy showed a 12-month relapse-free survival rate of 71%, supporting its potential role in prolonging remission.[34]

Table 5.1: Efficacy Results from the Pivotal Cohort of Study 2102-HEM-101

EndpointValueSource(s)
CR+CRh Rate (95% CI)35% (27-43%)6
CR Rate32%8
CRh Rate2.7%8
Median Time to CR+CRh1.9 months3
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 months3
Rate of Transfusion Independence34% (in baseline dependent patients)6

Safety Profile, Tolerability, and Management of Adverse Events

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.

Boxed Warning: Differentiation Syndrome (DS)

The most critical safety concern with Olutasidenib is Differentiation Syndrome, a risk highlighted by a boxed warning in the prescribing information.[8]

  • Incidence: In the pivotal trial, DS of any grade occurred in 14-16% of patients.[3] The events were often severe, with Grade 3 or higher DS occurring in 8-9% of patients. The syndrome was fatal in one patient (1%).[3]
  • Clinical Presentation: DS is a life-threatening, on-target toxicity resulting from the rapid proliferation and differentiation of myeloid cells. The clinical presentation is a systemic inflammatory response that can include fever, dyspnea, unexplained weight gain, pulmonary infiltrates, pleuropericardial effusions, hypotension, and acute kidney injury.[11] It is important to note that DS can occur with or without an accompanying elevation in the white blood cell count (leukocytosis).[36]
  • Timeline: The onset of DS is variable. It can occur as early as the first day of treatment or be delayed for up to 18 months.[14] The median time to onset in the clinical trial was 17.5 days, suggesting a period of high risk in the first few cycles of therapy.[3]
  • Management: Suspicion of DS requires immediate and decisive intervention. The cornerstone of management is the prompt initiation of systemic corticosteroids (e.g., dexamethasone 10 mg intravenously every 12 hours for a minimum of 3 days) and withholding Olutasidenib until symptoms resolve. If leukocytosis is present, treatment with a cytoreductive agent like hydroxyurea should be initiated. Close hemodynamic monitoring is essential.[7] With appropriate management, the majority of cases (76%) in the clinical trial recovered.[36]

Hepatotoxicity

Hepatotoxicity is another significant warning and precaution associated with Olutasidenib therapy.

  • Incidence: Liver-related adverse events are common. Hepatotoxicity of any grade was reported in 23-25% of patients, with Grade 3 or 4 events occurring in 13-15% of patients.[3]
  • Presentation: The toxicity typically manifests as asymptomatic elevations in liver function tests (LFTs), including alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase, and/or total bilirubin.[1]
  • Monitoring: Due to this risk, a rigorous LFT monitoring schedule is mandated. LFTs must be assessed prior to starting treatment, at least once weekly for the first two months, once every other week for the third month, once in the fourth month, and then once every other month for the duration of therapy.[10]
  • Management: Management of hepatotoxicity is based on the severity of the LFT elevations. It may require dose interruption, dose reduction to 150 mg once daily, or permanent discontinuation of the drug, as outlined in Table 6.2.[10]

Common and Serious Adverse Reactions

Beyond DS and hepatotoxicity, the overall safety profile of Olutasidenib is characterized by a range of common adverse events and hematologic toxicities.

  • Most Common (≥20%): The most frequently reported adverse reactions of any grade included nausea, fatigue/malaise, arthralgia (joint pain), constipation, leukocytosis, dyspnea (shortness of breath), fever, rash, mucositis (inflammation of mucous membranes), diarrhea, and transaminitis (elevated transaminases).[2]
  • Most Common Grade ≥3 TEAEs (≥10%): The most common severe (Grade 3 or 4) treatment-emergent adverse events were primarily hematologic, including febrile neutropenia (20-22%), anemia (20-22%), thrombocytopenia (low platelets, 16-28%), and neutropenia (low neutrophils, 13%).[3]

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 ReactionAll Grades (%)Grade 3-4 (%)Source(s)
Nausea38%0%8
Fatigue/Malaise36%3%8
Arthralgia28%3%8
Constipation26%0%8
Leukocytosis25%9%8
Dyspnea24%5%8
Fever (Pyrexia)24%1%8
Rash24%1%8
Mucositis23%3%8
Diarrhea20%1%8
Transaminitis20%12%8

Table 6.2: Guidelines for Dosage Modification in Response to Key Toxicities

ToxicityRecommended ActionSource(s)
Differentiation SyndromeWithhold 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 EffectsInterrupt 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

Dosage, Administration, and Drug Interactions

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.

Recommended Dosing Regimen and Formulation

  • Dosage: The recommended dosage of Olutasidenib is 150 mg taken orally twice daily (BID).[10]
  • Formulation: The drug is supplied as 150 mg opaque white hard gelatin capsules, which are imprinted with “OLU 150” for identification.[11]
  • Duration of Therapy: Treatment should be continued until there is evidence of disease progression or the development of unacceptable toxicity. Due to the drug's differentiation-based mechanism of action, a clinical response may take time to emerge. Therefore, it is recommended that patients without progression or unacceptable toxicity be treated for a minimum of 6 months to allow sufficient time for a response to occur.[11]

Administration Instructions

Patient education on proper administration is crucial to ensure consistent drug exposure and minimize risks.

  • Food Effect: As previously detailed, Olutasidenib must be taken on an empty stomach to avoid a significant increase in drug absorption and potential toxicity. Patients should be instructed to take their dose at least 1 hour before or 2 hours after a meal.[11]
  • Method of Administration: The capsules must be swallowed whole and should not be broken, opened, or chewed.[11]
  • Dosing Schedule: Doses should be administered at approximately the same times each day, and the two daily doses should not be taken within 8 hours of each other.[11]
  • Missed or Vomited Doses: If a dose is missed, the patient should take it as soon as possible, provided there are at least 8 hours until the next scheduled dose. They should then return to their normal schedule the following day. If a dose is vomited, a replacement dose should not be taken; the patient should wait until the next scheduled dose is due.[11]

Drug-Drug Interactions

Olutasidenib's metabolism and its own effect on metabolic enzymes create a complex profile of potential drug-drug interactions that must be carefully managed.

  • Effect of Other Drugs on Olutasidenib:
  • CYP3A Inducers: Olutasidenib is a major substrate of CYP3A4. Concomitant use with strong or moderate inducers of CYP3A4 (e.g., rifampin, carbamazepine, phenytoin, St. John's Wort) can significantly increase the metabolism of Olutasidenib, leading to lower plasma concentrations and a potential loss of efficacy. Therefore, coadministration with strong or moderate CYP3A inducers should be avoided.[10]
  • Effect of Olutasidenib on Other Drugs:
  • CYP3A4 Induction: Olutasidenib itself is a moderate inducer of CYP3A4.[3] This means it can accelerate the metabolism and reduce the plasma concentrations of other drugs that are sensitive CYP3A4 substrates. This could lead to a loss of therapeutic effect for the coadministered drug. Examples of sensitive CYP3A substrates include alfentanil, certain statins (e.g., simvastatin), and some benzodiazepines (e.g., midazolam).[10] Concomitant use should be avoided if possible. If unavoidable, the patient should be closely monitored for evidence of reduced efficacy of the substrate drug.[10]
  • Induction of Other CYPs: In addition to CYP3A4, Olutasidenib also induces several other CYP enzymes, including CYP2B6, CYP1A2, CYP2C8, and CYP2C9.[3] This broad induction profile creates the potential for a wide range of interactions with other medications.

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.

Regulatory Status and Clinical Development Landscape

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.

U.S. Food and Drug Administration (FDA)

  • Approval: The FDA granted regular approval to Olutasidenib on December 1, 2022.[1]
  • Indication: The approved indication is for the treatment of adult patients with relapsed or refractory acute myeloid leukemia (AML) with a susceptible isocitrate dehydrogenase-1 (IDH1) mutation as detected by an FDA-approved test.[11]
  • Companion Diagnostic: The approval of Olutasidenib was a landmark example of the precision oncology paradigm, as it occurred concurrently with the approval of its companion diagnostic, the Abbott RealTime IDH1 Assay.[8] This linkage is critical; the drug's indication is not for R/R AML in general, but is strictly limited to the molecularly-defined population in which it has proven effective. This regulatory approach ensures that treatment is inextricably tied to biomarker confirmation, maximizing the potential for benefit and avoiding use in patients who would not respond.
  • Regulatory Designations: During its development, Olutasidenib received an Orphan Drug designation from the FDA, acknowledging its development for a rare disease. Its approval was granted under a Standard Review process.[14]

European Medicines Agency (EMA)

  • Marketing Authorization: As of the latest available information, Olutasidenib has not received marketing authorization from the EMA and is not commercially available in the European Union.[34]
  • Orphan Designation: The EMA's Committee for Orphan Medicinal Products (COMP) granted Olutasidenib an orphan designation (EU/3/19/2159) on May 29, 2019, for the treatment of AML.[38] This designation is based on the seriousness and rarity of the condition and the preliminary evidence suggesting a potential for significant benefit over existing therapies. It is important to note that an orphan designation provides development incentives but is not equivalent to a marketing authorization.[38]
  • Pediatric Investigation Plan (PIP): The EMA granted a waiver for the requirement of a PIP in all pediatric age groups for the AML indication, concluding that the disease in children is fundamentally different or that the drug would not be effective or safe.[39]

Other Regulatory Agencies

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]

Ongoing and Future Clinical Trials

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.

  • Other Hematologic Malignancies: Olutasidenib is being investigated, both as a monotherapy and in combination with azacitidine, for patients with myelodysplastic syndrome (MDS) harboring an IDH1 mutation.[1]
  • Solid Tumors: Leveraging its ability to cross the blood-brain barrier, a key area of investigation is in mIDH1-positive solid tumors, particularly gliomas (NCT03684811).[1]
  • Expanded AML Settings: Research is ongoing to define the role of Olutasidenib in broader AML populations. This includes studies evaluating its use in combination with other agents like azacitidine and venetoclax, its potential as a frontline therapy for newly diagnosed patients, and its utility as a post-transplant maintenance therapy to prevent relapse.[7]

Comparative Analysis and Therapeutic Positioning

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.

Olutasidenib vs. Ivosidenib: A Cross-Trial Comparison

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]

  • Efficacy Comparison:
  • Response Rate: The rates of achieving a composite CR or CRh are very similar between the two agents. Olutasidenib demonstrated a CR+CRh rate of 35% in its pivotal cohort [6], while Ivosidenib's pivotal trial reported a CR+CRh rate of 32.8%.[14]
  • Duration of Response (DoR): This is the most significant point of differentiation. Olutasidenib has demonstrated a remarkably longer median duration of CR+CRh of 25.9 months. This is more than three times longer than the 8.2 months reported for Ivosidenib.[6] This profound difference in the durability of remission is Olutasidenib's key efficacy advantage.
  • Overall Survival (OS): The longer duration of response with Olutasidenib appears to translate into a survival benefit. Cross-trial comparisons of median OS favor Olutasidenib (11.6 months) over Ivosidenib (8.8 months).[7] Furthermore, a sophisticated indirect treatment comparison study using stabilized inverse probability of treatment weighting suggested a statistically significant overall survival advantage for Olutasidenib (Hazard Ratio: 0.33; 95% CI: 0.11, 0.94).[41]
  • Safety Profile Comparison:
  • Differentiation Syndrome: The incidence of the hallmark class effect, Differentiation Syndrome, is comparable between the two drugs, reported at 14-16% for Olutasidenib and 19% for Ivosidenib.[3]
  • Key Distinguishing Toxicities: The primary non-hematologic toxicities of concern are distinct. Olutasidenib is primarily associated with hepatotoxicity, with Grade 3 or higher events occurring in 13-15% of patients.[1] In contrast, the key toxicity for Ivosidenib is QTc interval prolongation, a cardiac-related side effect.[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.

Role in the Treatment Algorithm for mIDH1 R/R AML

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

FeatureOlutasidenib (Rezlidhia)Ivosidenib (Tibsovo)
CR+CRh Rate35%32.8%
Median Duration of CR+CRh25.9 months8.2 months
Median Overall Survival11.6 months8.8 months
Key Non-Hematologic ToxicityHepatotoxicityQTc Prolongation
Dosing Schedule150 mg twice daily (empty stomach)500 mg once daily (with or without food)
Source(s)33

Conclusion and Future Directions

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.

Summary of Olutasidenib's Risk-Benefit Profile

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.

Unanswered Questions and Areas for Future Research

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.

  • Combination Therapies: The most critical area of ongoing research is the evaluation of Olutasidenib in combination with other anti-leukemic agents. The goal is to improve upon the 35% response rate seen with monotherapy and to potentially prevent or overcome mechanisms of resistance. Early data for combinations with azacitidine are promising, and studies combining Olutasidenib with the BCL-2 inhibitor venetoclax are of high interest and are being actively pursued.[33]
  • Moving to Frontline Treatment: A logical next step is to investigate whether incorporating Olutasidenib into frontline treatment regimens can improve outcomes for newly diagnosed patients with mIDH1 AML, particularly those who are not candidates for intensive induction chemotherapy. Ongoing studies are exploring this question.[40]
  • Role as Maintenance Therapy: Given its oral administration and the durability of the responses it induces, Olutasidenib is an ideal candidate for maintenance therapy. Studies are underway to determine if its use after initial remission is achieved (either with chemotherapy or following hematopoietic stem cell transplantation) can prolong remission and improve long-term survival.[34]
  • Expansion to Other mIDH1 Cancers: The IDH1 mutation is a known driver in other cancers. The ability of Olutasidenib to penetrate the blood-brain barrier makes it a particularly attractive agent for mIDH1-mutated gliomas. Its potential in other mIDH1-driven solid tumors, such as cholangiocarcinoma and chondrosarcoma, also warrants and is undergoing clinical investigation.[1]

Final Perspective

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.

Works cited

  1. Olutasidenib | C18H15ClN4O2 | CID 118955396 - PubChem, accessed October 24, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Olutasidenib
  2. Olutasidenib - Wikipedia, accessed October 24, 2025, https://en.wikipedia.org/wiki/Olutasidenib
  3. Pharmacist's Application to Practice: Olutasidenib | HOPA, accessed October 24, 2025, https://www.hoparx.org/latest-news/pharmacists-application-to-practice-olutasidenib/
  4. Definition of olutasidenib - NCI Drug Dictionary - NCI, accessed October 24, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/olutasidenib
  5. What is the mechanism of Olutasidenib? - Patsnap Synapse, accessed October 24, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-olutasidenib
  6. FDA Approval Summary: Olutasidenib for Adult Patients with Relapsed or Refractory Acute Myeloid Leukemia with an Isocitrate Dehydrogenase-1 Mutation - PMC - PubMed Central, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11703669/
  7. Full article: Olutasidenib: a novel mutant IDH1 inhibitor for the treatment of relapsed or refractory acute myeloid leukemia - Taylor & Francis Online, accessed October 24, 2025, https://www.tandfonline.com/doi/full/10.1080/17474086.2024.2354486
  8. Rezlidhia FDA Approved for Relapsed or Refractory AML with IDH1 Mutation, accessed October 24, 2025, https://oncpracticemanagement.com/issues/2023/january-2023-vol-13-no-1/rezlidhia-fda-approved-for-relapsed-or-refractory-aml-with-idh1-mutation
  9. Olutasidenib (FT-2102) induces durable complete remissions in patients with relapsed or refractory IDH1-mutated AML - PubMed, accessed October 24, 2025, https://pubmed.ncbi.nlm.nih.gov/36724515/
  10. Rezlidhia (olutasidenib) dosing, indications, interactions, adverse effects, and more, accessed October 24, 2025, https://reference.medscape.com/drug/rezlidhia-olutasidenib-4000334
  11. Reference ID: 5086793 - accessdata.fda.gov, accessed October 24, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215814s000lbl.pdf
  12. FDA Approves Olutasidenib for Relapsed or Refractory AML with a Susceptible IDH1 Mutation - ACCC Cancer, accessed October 24, 2025, https://www.accc-cancer.org/detail-pages/blurb-discussion-detail/fda-approves-olutasidenib-for-relapsed-or-refractory-aml-with-a-susceptible-idh1-mutation
  13. FDA Approval Summary: Olutasidenib for Adult Patients with Relapsed or Refractory Acute Myeloid Leukemia with an Isocitrate Dehydrogenase 1 Mutation - PubMed, accessed October 24, 2025, https://pubmed.ncbi.nlm.nih.gov/39475462/
  14. Olutasidenib (Rezlidhia™) New Drug Update, accessed October 24, 2025, https://www.hhs.texas.gov/sites/default/files/documents/oct-2023-durb-agenda-item5z.pdf
  15. REZLIDHIA (olutasidenib), FDA-Approved for Treatment of Adult Patients with Relapsed or Refractory Acute Myeloid Leukemia, Available at Biologics by McKesson, accessed October 24, 2025, https://www.mckesson.com/about-us/newsroom/press-releases/2022/rezlidhia-fda-approved-treatment-relapsed-refractory-acute-myeloid-leukemia/
  16. Olutasidenib (FT-2102) | IDH1m Inhibitor | AmBeed.com, accessed October 24, 2025, https://www.ambeed.com/products/olutasidenib.html
  17. Olutasidenib | CAS 1887014-12-1 | Cayman Chemical | Biomol.com, accessed October 24, 2025, https://www.biomol.com/products/chemicals/biochemicals/olutasidenib-cay30370-1
  18. IDH1 Mutation | DrugBank Online, accessed October 24, 2025, https://go.drugbank.com/indications/DBCOND0119269
  19. 3D structure for Olutasidenib (DB16267) | DrugBank Online, accessed October 24, 2025, https://go.drugbank.com/structures/small_molecule_drugs/DB16267
  20. Olutasidenib | Dehydrogenase inhibitor | Mechanism | Concentration - Selleck Chemicals, accessed October 24, 2025, https://www.selleckchem.com/products/olutasidenib-ft-2102.html
  21. Olutasidenib (FT-2102, CAS Number: 1887014-12-1) | Cayman Chemical, accessed October 24, 2025, https://www.caymanchem.com/product/30370/olutasidenib
  22. Olutasidenib: from bench to bedside - PMC - PubMed Central, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10432604/
  23. Highlighting the mechanistic role of Olutasidenib (FT-2102) in the selective inhibition of mutated isocitrate dehydrogenase 1 (mIDH1) in cancer therapy | Request PDF - ResearchGate, accessed October 24, 2025, https://www.researchgate.net/publication/357228959_Highlighting_the_mechanistic_role_of_Olutasidenib_FT-2102_in_the_selective_inhibition_of_mutated_isocitrate_dehydrogenase_1_mIDH1_in_cancer_therapy
  24. Olutasidenib (FT-2102) induces durable complete remissions in patients with relapsed or refractory IDH1-mutated AML - PubMed Central, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10362540/
  25. Olutasidenib (FT-2102) | IDH1m Inhibitor | MedChemExpress, accessed October 24, 2025, https://www.medchemexpress.com/Olutasidenib.html
  26. Olutasidenib: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 24, 2025, https://go.drugbank.com/drugs/DB16267
  27. Olutasidenib: uses, dosing, warnings, adverse events, interactions - Oncology News Central, accessed October 24, 2025, https://www.oncologynewscentral.com/drugs/monograph/185475-322057/olutasidenib-oral
  28. olutasidenib | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed October 24, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=clinical&ligandId=10319
  29. Open-label Study of FT-2102 With or Without Azacitidine or Cytarabine in Patients With AML or MDS With an IDH1 Mutation - larvol clin, accessed October 24, 2025, https://clin.larvol.com/trial-detail/NCT02719574
  30. FDA Approves Treatment for R/R AML | ASH Clinical News | American Society of Hematology, accessed October 24, 2025, https://ashpublications.org/ashclinicalnews/news/6956/FDA-Approves-Treatment-for-R-R-AML
  31. FDA D.I.S.C.O. Burst Edition: FDA approval of Rezlidhia ..., accessed October 24, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-disco-burst-edition-fda-approval-rezlidhia-olutasidenib-relapsed-or-refractory-acute-myeloid
  32. Olutasidenib alone or with azacitidine in IDH1-mutated acute myeloid leukaemia and myelodysplastic syndrome: phase 1 results of a phase 1/2 trial | Request PDF - ResearchGate, accessed October 24, 2025, https://www.researchgate.net/publication/365289503_Olutasidenib_alone_or_with_azacitidine_in_IDH1-mutated_acute_myeloid_leukaemia_and_myelodysplastic_syndrome_phase_1_results_of_a_phase_12_trial
  33. Olutasidenib alone or with azacitidine in IDH1-mutated acute ..., accessed October 24, 2025, https://www.thelancet.com/journals/lanhae/article/PIIS2352-3026(22)00292-7/fulltext
  34. Olutasidenib | MedPath, accessed October 24, 2025, https://trial.medpath.com/drug/0352771e30deecfa/olutasidenib
  35. Olutasidenib Maintenance Generates Durable RFS in IDH1-Mutated AML Following Induction CR/CRi | OncLive, accessed October 24, 2025, https://www.onclive.com/view/olutasidenib-maintenance-generates-durable-rfs-in-idh1-mutated-aml-following-induction-cr-cri
  36. Olutasidenib | American Journal of Health-System Pharmacy | Oxford Academic, accessed October 24, 2025, https://academic.oup.com/ajhp/article/80/7/398/7035018
  37. New Medicines Approved in 2022 - Meds Entry Watch - Canada.ca, accessed October 24, 2025, https://www.canada.ca/en/patented-medicine-prices-review/services/npduis/analytical-studies/meds-entry-watch-8th-edition/new-medicines-approved-2022.html
  38. EU/3/19/2159 - orphan designation for treatment of acute myeloid ..., accessed October 24, 2025, https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu-3-19-2159
  39. EMEA-003421-PIP01-23 - paediatric investigation plan - European Medicines Agency, accessed October 24, 2025, https://www.ema.europa.eu/en/medicines/human/paediatric-investigation-plans/emea-003421-pip01-23
  40. Rigel Announces Second REZLIDHIA® (Olutasidenib) Publication in Blood Advances, accessed October 24, 2025, https://www.rigel.com/news-media/press-releases/detail/362/rigel-announces-second-rezlidhia-olutasidenib
  41. Full article: Effectiveness of olutasidenib versus ivosidenib in patients with mutated isocitrate dehydrogenase 1 acute myeloid leukemia who are relapsed or refractory to venetoclax: the 2102-HEM-101 trial versus a US electronic health record-based external control arm - Taylor & Francis Online, accessed October 24, 2025, https://www.tandfonline.com/doi/full/10.1080/10428194.2025.2514894?af=R

Published at: October 24, 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.