Report on Ziftomenib (KO-539): A Novel Menin Inhibitor for Acute Myeloid Leukemia
I. Introduction and Executive Summary
Ziftomenib, also known by its developmental code name KO-539, is an orally bioavailable, investigational small molecule drug poised to introduce a new paradigm in the treatment of specific, genetically defined subsets of Acute Myeloid Leukemia (AML).[1] As a member of the novel therapeutic class of menin inhibitors, Ziftomenib employs a highly targeted mechanism of action, designed to disrupt a critical protein-protein interaction that drives cancer cell proliferation and survival in certain leukemias.[3]
The clinical development of Ziftomenib is precisely focused on AML patient populations with a profound unmet medical need: those whose disease is characterized by mutations in the nucleophosmin 1 (NPM1) gene or rearrangements of the lysine methyltransferase 2A (KMT2A, formerly known as MLL) gene.[3] These genetic alterations are found in a significant portion of AML cases, and for patients who relapse or are refractory to initial therapies, the prognosis is exceptionally poor, with limited effective treatment options currently available.[6]
The comprehensive clinical program for Ziftomenib is anchored by two key studies. The KOMET-001 trial, a Phase 1/2 study, evaluated Ziftomenib as a monotherapy in patients with relapsed/refractory (R/R) AML. This trial successfully identified a recommended Phase 2 dose and, most critically, demonstrated significant and durable clinical activity in the NPM1-mutant cohort, providing the pivotal data supporting a New Drug Application (NDA) to the U.S. Food and Drug Administration (FDA).[9] The KOMET-007 trial is exploring Ziftomenib in the frontline setting, where its combination with standard-of-care chemotherapy has yielded exceptionally high rates of deep, molecular remissions in newly diagnosed patients with both
NPM1 mutations and KMT2A rearrangements.[11]
Developed through a strategic global collaboration between Kura Oncology, Inc. and Kyowa Kirin Co., Ltd., Ziftomenib is on an accelerated regulatory trajectory.[6] In June 2025, the FDA accepted the NDA for Ziftomenib for the treatment of adult patients with R/R
NPM1-mutant AML, granting it Priority Review and assigning a Prescription Drug User Fee Act (PDUFA) target action date of November 30, 2025.[9] This follows the receipt of multiple expedited designations, including Breakthrough Therapy, Fast Track, and Orphan Drug status, underscoring the agent's potential to provide a substantial improvement over existing therapies.[6] If approved, Ziftomenib is positioned to become the first menin inhibitor and the first targeted therapy specifically for the R/R
NPM1-mutant AML population.[14]
In summary, Ziftomenib represents a significant and promising advance in the field of precision oncology for AML. Its targeted mechanism, supported by a favorable pharmacokinetic profile and compelling clinical data demonstrating a positive benefit-risk profile, establishes a clear path toward regulatory approval and a transformative role in the management of this challenging disease.
II. Molecular Profile and Physicochemical Characteristics
A precise understanding of a drug's molecular identity and properties is fundamental to its development, formulation, and regulatory evaluation. Ziftomenib is a structurally complex synthetic organic compound classified as a small molecule drug.[1]
Drug Identification
The compound is recognized globally by several key identifiers:
- Generic Name: Ziftomenib, as assigned by the United States Adopted Names (USAN) Council and as an International Nonproprietary Name (INN).[1]
- Developmental Code Name: KO-539, used during its preclinical and early clinical development phases.[1]
- Drug Type: Small Molecule.[1]
Chemical and Structural Information
Ziftomenib's precise chemical structure is defined by its formal nomenclature and various machine-readable formats.
- IUPAC Name: 4-methyl-5-[[[2-(methylamino)-6-(2,2,2-trifluoroethyl)thieno[2,3-d]pyrimidin-4-yl]amino]piperidin-1-yl]methyl]-1-indole-2-carbonitrile.[1]
- Molecular Formula: C33H42F3N9O2S2.[1]
- Molecular Weight: Values are consistently reported in the range of 717.87 to 717.9 g/mol.[1]
- Structural Representations:
- SMILES: CC1=C(C=CC2=C1C=C(N2C[C@H](C)N3CCN(CC3)S(=O)(=O)C)C#N)CN4CCC(CC4)NC5=C6C=C(SC6=NC(=N5)NC)CC(F)(F)F.[1]
- InChI: InChI=1S/C33H42F3N9O2S2/c1-21(43-11-13-44(14-12-43)49(4,46)47)19-45-25(18-37)15-27-22(2)23(5-6-29(27)45)20-42-9-7-24(8-10-42)39-30-28-16-26(17-33(34,35)36)48-31(28)41-32(38-3)40-30/h5-6,15-16,21,24H,7-14,17,19-20H2,1-4H3,(H2,38,39,40,41)/t21-/m0/s1.[1]
- InChIKey: BGGALFIXXQOTPY-NRFANRHFSA-N.[1]
Database and Regulatory Identifiers
For unambiguous cross-referencing across global scientific and regulatory databases, Ziftomenib is assigned a comprehensive set of unique identifiers. These identifiers, along with key computed physicochemical properties that predict its drug-like behavior, are consolidated in Table 1. The properties listed, such as a molecular weight exceeding 500 Da and a high number of hydrogen bond acceptors, result in one violation of Lipinski's rule of five, a common characteristic of complex modern therapeutics designed to inhibit challenging targets like protein-protein interactions.[4]
Table 1: Key Identifiers and Physicochemical Properties of Ziftomenib
| Identifier Type | Value | Source(s) |
|---|
| Chemical Identifiers | | |
| CAS Number | 2134675-36-6 | 1 |
| Molecular Formula | C33H42F3N9O2S2 | 1 |
| Molecular Weight | 717.88 g/mol | 2 |
| Database Identifiers | | |
| DrugBank ID | DB17171 | 1 |
| PubChem CID | 138497449 | 5 |
| UNII (FDA GSRS) | 4MOD1F4ENC | 1 |
| ChEMBL ID | CHEMBL5095038 | 1 |
| KEGG ID | D12419 | 1 |
| NCI Thesaurus Code | C164227 | 1 |
| PDB Ligand | K5O | 5 |
| Computed Physicochemical Properties | | |
| XLogP | 4.73 - 5.6 | 1 |
| Hydrogen Bond Donors | 2 | 1 |
| Hydrogen Bond Acceptors | 10 - 14 | 1 |
| Rotatable Bonds | 10 - 11 | 4 |
| Topological Polar Surface Area (TPSA) | 159 Ų | 1 |
This consolidation of identifiers and properties provides a definitive molecular foundation, essential for accurate data retrieval, bioinformatic analysis, and regulatory documentation. It confirms Ziftomenib's identity as a single, well-characterized chemical entity and offers preliminary insights into its biopharmaceutical characteristics, such as its considerable size and polarity, which influence its absorption and distribution.
III. Preclinical and Clinical Pharmacology
The pharmacological profile of Ziftomenib is defined by its highly specific mechanism of action (pharmacodynamics) and its behavior within the human body (pharmacokinetics). Together, these characteristics provide the scientific rationale for its clinical use, dosing regimen, and targeted patient population.
A. Mechanism of Action (Pharmacodynamics)
Ziftomenib's therapeutic effect is rooted in its ability to disrupt a fundamental oncogenic process in specific AML subtypes.
The Menin-KMT2A Axis in Leukemogenesis
In normal hematopoiesis, the protein menin acts as a critical scaffolding component of a larger protein complex involving lysine methyltransferase 2A (KMT2A).[18] This menin-KMT2A complex is essential for regulating the transcription of key developmental genes, particularly those in the
HOX and MEIS1 families, which control cell differentiation and lineage commitment.[20]
In two specific subtypes of AML, this regulatory axis is hijacked to drive cancer:
- KMT2A-rearranged (KMT2A-r) AML: Chromosomal translocations involving the KMT2A gene create fusion proteins. These fusion proteins aberrantly recruit the menin-containing complex to target genes, leading to their sustained overexpression, which blocks hematopoietic differentiation and promotes the uncontrolled proliferation of leukemic blasts.[8]
- NPM1-mutated (NPM1-m) AML: Mutations in NPM1 cause the protein to be abnormally localized in the cytoplasm. This disrupts normal cellular processes and, through a distinct but functionally convergent mechanism, also leads to a dependency on the wild-type menin-KMT2A interaction to maintain the expression of the same leukemogenic genes (HOX, MEIS1).[7]
In both KMT2A-r and NPM1-m AML, the interaction between menin and KMT2A (or its fusion product) is an absolute requirement for the survival and proliferation of the cancer cells, making it a prime therapeutic target.[1]
Ziftomenib's Molecular Target
Ziftomenib is a potent, selective, and orally active inhibitor of the menin-KMT2A protein-protein interaction (PPI).[1] It functions by binding directly to a pocket on the menin protein, physically preventing it from associating with KMT2A.[1] This targeted disruption is the core of its mechanism. The development of an effective small molecule to inhibit a PPI, which involves large and often featureless binding surfaces, represents a significant achievement in medicinal chemistry.
Downstream Consequences
By breaking the critical menin-KMT2A link, Ziftomenib initiates a cascade of anti-leukemic events. The aberrant transcriptional program is shut down, leading to a marked reduction in the expression of key downstream oncogenes, including MEIS1, PBX3, FLT3, and the anti-apoptotic protein BCL2.[1] This transcriptional reprogramming lifts the differentiation block that characterizes AML. As a result, the cancerous juvenile blast cells are induced to mature into healthy white blood cells or, alternatively, are pushed toward programmed cell death (apoptosis).[3] The ultimate outcome is a potent inhibition of the proliferation and survival of the leukemic cells that are dependent on this pathway.[2] This high degree of specificity for cells with
KMT2A rearrangements or NPM1 mutations explains why Ziftomenib is a quintessential precision medicine, effective only in patients whose tumors possess the targetable biological vulnerability.
B. Pharmacokinetic (PK) Profile (ADME)
A Phase 1 study in healthy male volunteers has characterized the absorption, distribution, metabolism, and excretion (ADME) properties of Ziftomenib, providing a clear understanding of its behavior in the body and supporting its clinical dosing strategy.[25]
- Absorption: Following oral administration, Ziftomenib is rapidly absorbed into the bloodstream. The median time to reach maximum plasma concentration (Tmax) is 3.5 hours.[25]
- Distribution: While specific tissue distribution data is not detailed, the drug's oral bioavailability indicates it achieves systemic circulation to exert its effect on leukemic cells in the bone marrow and blood.
- Metabolism: Ziftomenib undergoes limited metabolism after absorption. The primary metabolic pathways identified are oxidation, N-demethylation, and N-dealkylation. Although 19 metabolites were recovered in plasma, all were present at low levels, constituting less than 10% of the total drug-related exposure. The unchanged parent drug, Ziftomenib, is the most abundant component in plasma, indicating that it is the primary active moiety and is not extensively broken down by the liver.[25]
- Excretion: The elimination of Ziftomenib and its metabolites occurs predominantly through the fecal route. In a mass balance study using radiolabeled drug, 89.7% of the total radioactivity was recovered in feces over a 480-hour period, while a negligible 0.5% was found in urine. This indicates that the drug is primarily cleared as unchanged parent drug via biliary or direct intestinal excretion.[25]
- Bioavailability and Half-life: The absolute oral bioavailability of Ziftomenib was determined to be 12.9%. A key feature of its PK profile is its long elimination half-life (t1/2), which was measured at 61.5 hours. This prolonged duration of action is a critical characteristic that strongly supports the feasibility and convenience of a once-daily (QD) dosing regimen.[25]
The pharmacokinetic properties of Ziftomenib are summarized in Table 2. The combination of oral administration, rapid absorption, and a long half-life creates a highly favorable profile for clinical use. This profile simplifies the dosing schedule for patients, enhancing convenience and likely improving treatment adherence, particularly in the context of long-term or maintenance therapy. The predominantly fecal route of elimination also suggests a lower risk of drug accumulation in patients with renal impairment, although this would require specific clinical evaluation.
Table 2: Summary of Pharmacokinetic Parameters of Ziftomenib in Healthy Volunteers
| PK Parameter | Value | Clinical Implication | Source(s) |
|---|
| Administration Route | Oral | Patient convenience, suitable for outpatient use | 1 |
| Time to Max Concentration (Tmax) | 3.5 hours (median) | Rapid absorption after dosing | 25 |
| Elimination Half-life (t1/2) | 61.5 hours | Supports a convenient once-daily (QD) dosing schedule | 25 |
| Absolute Bioavailability | 12.9% | Sufficient oral absorption to achieve therapeutic concentrations | 25 |
| Primary Route of Excretion | Fecal (89.7%) | Minimal renal clearance, suggesting lower risk in renal impairment | 25 |
| Metabolism | Limited; parent drug is major component in plasma | Parent drug is the primary active agent; low potential for major active metabolites | 25 |
IV. Clinical Efficacy in Relapsed/Refractory Acute Myeloid Leukemia (KOMET-001 Trial)
The KOMET-001 study (NCT04067336) is the cornerstone clinical trial that established the efficacy and safety of Ziftomenib monotherapy, providing the pivotal evidence for its NDA submission.[9] This multicenter, open-label, Phase 1/2 trial was designed to evaluate the drug in heavily pretreated adult patients with R/R AML, a population with a grim prognosis and few effective therapeutic options.[5]
Trial Design and Population
The study consisted of several phases. Phase 1a was a dose-escalation phase open to patients with all molecular subtypes of AML, with the primary goal of determining the maximum tolerated dose and recommended Phase 2 dose (RP2D). Phase 1b was a dose-validation and expansion phase that specifically enrolled patients with the target genetic alterations: NPM1 mutations or KMT2A rearrangements. Patients in this phase were randomized to receive either 200 mg or 600 mg of Ziftomenib orally once daily in 28-day cycles.[9] The trial population was characteristic of R/R AML, having received multiple prior lines of therapy.[10]
Dose Finding and Recommended Phase 2 Dose (RP2D)
The dose-escalation and validation phases clearly demonstrated a dose-response relationship. While the 200 mg dose level showed minimal activity, with no responses reported in the Phase 1b cohort, the 600 mg dose emerged as superior.[10] Based on an optimal balance of clinical activity and a manageable safety profile, 600 mg once daily was selected as the RP2D for the pivotal Phase 2 portion of the trial in patients with
NPM1-mutant AML.[10]
Efficacy in NPM1-Mutant R/R AML
The performance of Ziftomenib in the NPM1-mutant cohort at the 600 mg RP2D represents the primary success of the monotherapy program and the foundation of the NDA. In this population, the KOMET-001 trial successfully met its primary endpoint, demonstrating a statistically significant rate of complete remission (CR) plus CR with partial hematologic recovery (CRh).[9]
- Response Rates: The clinical activity observed was both significant and meaningful. In a pooled analysis of the Phase 1b/2 cohorts treated at 600 mg, the key response rates were as follows:
- CR/CRh Rate: 25% (28 out of 112 patients).[29] An earlier report from a smaller cohort of 20 patients noted a 30% CR rate (6 patients) and a 35% CRc rate.[26]
- Composite CR (CRc) Rate: 29% (32 out of 112 patients).[29]
- Overall Response Rate (ORR): 40%.[30]
- Durability and Depth of Response: The quality of the responses was a key finding.
- Duration: The remissions were durable, with a median duration of response (DoR) for patients achieving CRc estimated at 8.2 months.[30]
- Depth: A high proportion of responding patients achieved measurable residual disease (MRD) negativity, a critical indicator of a deep molecular remission that is associated with improved long-term outcomes. Reports indicate that 65% to 67% of evaluable responders were MRD-negative.[26]
- Clinical Impact: The remissions were clinically meaningful, as several patients who responded to Ziftomenib were successfully bridged to a potentially curative allogeneic hematopoietic stem cell transplant (HSCT).[30]
- Subgroup Analysis: Importantly, responses were observed across various subgroups, including patients with prior exposure to the BCL-2 inhibitor venetoclax and those with common co-mutations such as FLT3 and IDH, which can confer a worse prognosis.[26]
Efficacy in KMT2A-Rearranged R/R AML
In contrast to the NPM1-mutant cohort, Ziftomenib monotherapy demonstrated more modest clinical activity in patients with KMT2A rearrangements.[10] While some responses were observed, the overall efficacy was lower, and this, combined with a more challenging safety profile in this specific subgroup, led to a pivotal strategic decision. The developers halted further enrollment of
KMT2A-rearranged patients into the monotherapy arms of the trial to instead pursue a more promising combination therapy approach for this population.[10] This adaptation based on emerging data highlights a sophisticated approach to clinical development, redirecting resources toward the strategy with the highest probability of success for each distinct patient population.
The key efficacy outcomes for the successful NPM1-mutant R/R AML cohort are summarized in Table 3. This data established a new benchmark for a targeted agent in a patient population with no approved targeted therapies and a median overall survival of only a few months with standard salvage treatments.[6] The ability to induce deep, durable, and MRD-negative remissions provides a powerful rationale for its approval and adoption into clinical practice.
Table 3: Efficacy Outcomes of Ziftomenib Monotherapy in R/R NPM1-mutant AML (KOMET-001, 600 mg Dose)
| Efficacy Endpoint | Rate (%) | Median Duration (months) | Key Details | Source(s) |
|---|
| CR + CRh | 25% | 3.7 | Primary endpoint of the registrational trial; statistically significant. | 9 |
| Composite CR (CRc) | 29% | 8.2 | Includes CR, CRh, and CR with incomplete recovery. | 29 |
| Overall Response Rate (ORR) | 35% - 40% | Not Reported | Includes partial responses and morphologic leukemia-free state. | 30 |
| MRD Negativity in Responders | 65% - 67% | Not Applicable | Indicates deep molecular remission in a majority of responders. | 26 |
| Bridge to Transplant | Achieved | Not Applicable | Several responding patients were able to proceed to potentially curative HSCT. | 30 |
V. Clinical Efficacy in the Frontline Setting: Combination Therapy (KOMET-007 Trial)
Following the success of monotherapy in the R/R setting, the clinical development program for Ziftomenib logically advanced into the frontline setting with the KOMET-007 trial (NCT05735184). This ongoing Phase 1a/b study is designed to evaluate the safety and efficacy of Ziftomenib when added to the standard-of-care intensive induction chemotherapy regimen (a combination of cytarabine and an anthracycline like daunorubicin, commonly known as "7+3").[11] The trial enrolls newly diagnosed (ND) adult patients with AML harboring either
NPM1 mutations or KMT2A rearrangements. The central hypothesis is that introducing a highly active targeted agent at the time of initial diagnosis can deepen the response to chemotherapy, eradicate residual disease, and ultimately improve long-term outcomes and cure rates.
Impressive Efficacy in Combination
Interim results from the KOMET-007 trial have demonstrated robust and profound clinical activity, suggesting a powerful synergistic or additive effect between Ziftomenib and intensive chemotherapy. The data, particularly from the 600 mg Ziftomenib dose cohort, have been exceptionally strong.[12]
- Response Rates: Among response-evaluable patients, the rates of remission were remarkably high across both genetic subtypes.
- Overall Composite Complete Remission (CRc): An impressive 92% (65 out of 71) of patients across both genotypes achieved a CRc.[12]
- NPM1-mutant Cohort: This group showed near-universal responses, with a 93% CRc rate (41 out of 44 patients) and an 84% CR rate.[12]
- KMT2A-rearranged Cohort: This population, which showed only modest benefit from monotherapy, experienced a dramatic improvement with the combination therapy. The trial reported an 89% CRc rate (24 out of 27 patients) and a 74% CR rate.[12] This finding strongly validates the strategic decision to pivot this subgroup to a combination approach.
- Depth and Speed of Response: Beyond the high remission rates, the quality of these remissions is a critical finding.
- MRD Negativity: The combination therapy led to very high rates of MRD negativity, a key predictor of long-term survival. Among patients who achieved a CR, 71% of those with NPM1 mutations and 88% of those with KMT2A rearrangements became MRD-negative.[12]
- Rapidity: These deep molecular responses were achieved quickly, with a median time to MRD negativity of approximately 4.4 to 4.7 weeks.[12]
The striking efficacy of the combination therapy is summarized in Table 4. These results position Ziftomenib as a potentially transformative addition to the frontline standard of care. Achieving such high rates of MRD-negative remissions so rapidly suggests that the combination is highly effective at eradicating the leukemic clone, which is the fundamental goal of induction therapy. This level of activity provides a strong rationale for the planned Phase 3 registration-directed trials (the KOMET-017 program), which will be designed to confirm these findings and formally establish Ziftomenib as a new standard of care in the frontline setting.[12]
Table 4: Efficacy Outcomes of Ziftomenib in Combination with 7+3 in Newly Diagnosed AML (KOMET-007)
| Patient Cohort | N (Response-Evaluable) | CRc Rate (%) | CR Rate (%) | MRD Negativity Rate (in CR patients) (%) | Source(s) |
|---|
| NPM1-mutant | 44 | 93% | 84% | 71% | 12 |
| KMT2A-rearranged | 27 | 89% | 74% | 88% | 12 |
| Overall | 71 | 92% | 80% | Not Reported | 12 |
VI. Comprehensive Safety and Tolerability Assessment
A thorough evaluation of a drug's safety profile is essential to determine its overall benefit-risk balance. Across its clinical development program, Ziftomenib has demonstrated a generally well-tolerated and manageable safety profile, with consistent findings across studies.[9] A key feature is the low rate of treatment discontinuations due to drug-related adverse events, reported at just 3% in the pivotal KOMET-001 trial, indicating that most toxicities can be effectively managed without forcing patients to stop a potentially beneficial therapy.[15]
Common Treatment-Emergent Adverse Events (TEAEs)
The most frequently reported Grade 3 or higher TEAEs are largely consistent with those expected in a patient population with AML undergoing treatment. These primarily include:
- Hematologic Toxicities: Anemia, thrombocytopenia (low platelet count), and febrile neutropenia (fever with low white blood cell count) are common.[10]
- Infections: Pneumonia and sepsis are significant risks for immunocompromised AML patients and were reported in the trials.[10]
When Ziftomenib was combined with "7+3" intensive chemotherapy in the KOMET-007 trial, the safety profile remained consistent with that of intensive chemotherapy alone. Crucially, the addition of Ziftomenib did not appear to cause additional myelosuppression (suppression of bone marrow function) or delay the time to neutrophil and platelet recovery, a critical safety consideration for combination regimens.[11]
Adverse Event of Special Interest: Differentiation Syndrome (DS)
The most notable and clinically significant adverse event associated with Ziftomenib is Differentiation Syndrome (DS). This is a known on-target toxicity for agents that induce the maturation of leukemic cells.
- Mechanism and Presentation: DS is a systemic inflammatory response triggered by the rapid differentiation of a large burden of leukemic blasts. It can manifest with symptoms such as fever, respiratory distress, fluid accumulation (pleural/pericardial effusions, peripheral edema), hypotension, and acute renal failure.[23]
- Incidence and Severity: The incidence and severity of DS were closely monitored and proved to be a key factor in shaping the drug's development strategy.
- In the KOMET-001 monotherapy trial, any-grade DS was reported in 13% to 20% of patients.[10] Grade 3 or higher events occurred in 5% to 15% of patients.[10]
- A critical observation was that the incidence and severity of DS appeared to be higher in the KMT2A-rearranged cohort. This culminated in a treatment-related death attributed to DS in this subgroup, which was a primary driver of the decision to halt monotherapy development for these patients and pivot to a combination strategy.[10]
- Management: Despite its potential severity, DS is a manageable condition with well-established protocols. Early recognition is key, and management typically involves the prompt administration of corticosteroids (e.g., dexamethasone) and, if necessary, temporary interruption of Ziftomenib. With proactive management, most DS events were reported to resolve, and later analyses of the pivotal trial noted no Grade 4 or 5 events, indicating that clinical teams became adept at controlling this toxicity.[31]
The safety profile of Ziftomenib is distinct from that of its main competitor, revumenib, which is associated with a significant risk of QTc interval prolongation (an electrical disturbance in the heart).[23] In contrast, QTc prolongation has been reported only infrequently and at low grades with Ziftomenib (e.g., 2% Grade 3), suggesting a more favorable cardiac safety profile.[11] This lack of a significant cardiac signal could become a key differentiating factor in clinical practice.
Table 5 provides a summary of the most common severe adverse events observed in both the monotherapy and combination therapy settings.
Table 5: Summary of Common (≥10%) Grade ≥3 Treatment-Emergent Adverse Events
| Adverse Event | Monotherapy (KOMET-001, %) | Combination (KOMET-007, %) | Key Management Notes | Source(s) |
|---|
| Differentiation Syndrome | 13% - 15% | Not specified, but no new signals | Key on-target toxicity; managed with steroids and dose interruption. Higher severity in KMT2A-r monotherapy. | 10 |
| Febrile Neutropenia | 22% | 56% (TRAE: 15%) | Common with AML therapy; managed with antibiotics and supportive care. | 10 |
| Anemia | 21% - 25% | 32% (TRAE: 11%) | Managed with transfusions as needed. | 10 |
| Thrombocytopenia | 13% - 20% | 47% (TRAE: 15%) | Managed with platelet transfusions as needed. | 10 |
| Pneumonia | 16% - 20% | Not specified | A common and serious infection in this population. | 10 |
| Sepsis | 12% | Not specified | A life-threatening infection requiring urgent medical care. | 10 |
VII. Regulatory and Development Trajectory
The path of Ziftomenib from an early-stage clinical candidate to a drug on the verge of potential market approval has been shaped by a strategic collaboration, an efficient regulatory strategy, and ambitious plans for future development.
Sponsors and Collaboration
Ziftomenib is being developed by Kura Oncology, a clinical-stage biopharmaceutical company focused on precision medicines for cancer.[26] In a significant strategic move in November 2024, Kura Oncology entered into a global collaboration agreement with Kyowa Kirin, a well-established Japanese pharmaceutical company.[6] This partnership is comprehensive, covering the co-development and co-commercialization of Ziftomenib for AML and other hematologic malignancies. The collaboration provides Kura with substantial financial backing—evidenced by a $45 million milestone payment triggered by the NDA submission—as well as access to Kyowa Kirin's global infrastructure and expertise, which will be crucial for a successful worldwide launch and market penetration.[6]
U.S. FDA Pathway
Kura Oncology has masterfully navigated the regulatory process with the U.S. FDA, leveraging all available expedited programs to accelerate the development and review of Ziftomenib. This demonstrates the agency's recognition of the drug's potential to address a serious unmet medical need.
- Expedited Designations: For the indication of R/R NPM1-mutant AML, Ziftomenib has been granted a full suite of designations:
- Orphan Drug Designation: Provides market exclusivity and financial incentives for the development of drugs for rare diseases.[6]
- Fast Track Designation: Facilitates more frequent interactions with the FDA to expedite development and review.[6]
- Breakthrough Therapy Designation: Granted in April 2024, this is reserved for drugs that may demonstrate substantial improvement over available therapy on a clinically significant endpoint. Ziftomenib is the first and only investigational agent to receive this designation for R/R NPM1-mutant AML, highlighting its perceived therapeutic potential.[5]
- New Drug Application (NDA) Submission and Review:
- Building on the positive results of the KOMET-001 trial, Kura Oncology submitted the NDA to the FDA on March 31, 2025, seeking full approval for Ziftomenib in adult patients with R/R NPM1-mutant AML.[6]
- In June 2025, the FDA accepted the NDA for filing and, importantly, granted it Priority Review. This designation shortens the review timeline from the standard 10 months to 6 months and is reserved for drugs that could provide significant improvements in the safety or effectiveness of the treatment of serious conditions.[9]
- The FDA has assigned a PDUFA target action date of November 30, 2025, by which it is expected to make a decision on the approval of Ziftomenib.[9]
European EMA Pathway
Ziftomenib is also advancing in Europe. On January 12, 2024, the European Medicines Agency (EMA) granted it orphan medicine designation for the treatment of AML. This status provides scientific and regulatory support to facilitate its development and review in the European Union.[34]
Future Development Plans
The development strategy for Ziftomenib extends far beyond its initial indication. The company is actively pursuing a broad clinical program to establish its role across the AML treatment continuum. The cornerstone of this future strategy is the KOMET-017 program. This global protocol encompasses two independent, randomized, double-blind, placebo-controlled Phase 3 trials designed to evaluate Ziftomenib in combination with standards of care for newly diagnosed NPM1-mutant or KMT2A-rearranged AML. These trials, which will assess Ziftomenib with both intensive and non-intensive chemotherapy backbones, are intended to provide the definitive evidence needed to expand its label into the larger frontline market.[12]
VIII. Therapeutic Positioning and Strategic Analysis
Ziftomenib is entering a dynamic therapeutic landscape for AML. Its success will depend not only on its clinical merits but also on the severity of the unmet medical need it addresses and its competitive positioning against other emerging therapies.
A. The Unmet Need and Current Standard of Care (SoC)
Ziftomenib targets two patient populations with distinct but significant needs.
- NPM1-mutant AML: This is the most common genetic alteration in AML, occurring in approximately 30% of patients.[7] In the frontline setting, patients without a concurrent
FLT3-ITD mutation are often considered to have a favorable risk profile and respond well to standard "7+3" induction chemotherapy, with high initial remission rates.[36] However, a substantial portion of these patients will eventually relapse.[6] In the
relapsed/refractory setting, the prognosis is dismal. There are currently no FDA-approved targeted therapies specifically for NPM1-mutant AML.[6] Treatment relies on salvage chemotherapy or venetoclax-based combinations, which have limited efficacy and durability in this context, leading to a median overall survival of only a few months.[6] Ziftomenib is therefore positioned to fill a critical therapeutic void for these patients.
- KMT2A-rearranged AML: While less common, occurring in 5-10% of adult AML cases, KMT2A rearrangements are more prevalent in pediatric and therapy-related AML and are associated with a particularly aggressive disease course.[23] These patients have historically had a poor prognosis with high rates of relapse, even after intensive chemotherapy and allogeneic stem cell transplant.[8] The development of targeted therapies for this subgroup is a high priority.
B. Competitive Landscape: The Race of Menin Inhibitors
Ziftomenib is a leader in a new class of drugs, but it is not alone. The primary competition comes from another advanced-stage menin inhibitor, creating a dynamic "race to market" and setting the stage for a competitive duopoly.
- Primary Competitor: Revumenib (Syndax Pharmaceuticals):
- Mechanism: Revumenib (formerly SNDX-5613) shares the same mechanism of action as Ziftomenib, inhibiting the menin-KMT2A interaction to target the same NPM1-mutant and KMT2A-rearranged leukemias.[23]
- Development Status: Revumenib is also at a very advanced stage of development. Its pivotal trial, AUGMENT-101, has produced positive results, and an NDA for the treatment of R/R KMT2A-rearranged acute leukemia is under review by the FDA, with a potential approval decision anticipated in late 2024. This positions revumenib to potentially reach the market slightly ahead of Ziftomenib, but for a different lead indication.[21]
- Efficacy Comparison: Data from their respective pivotal trials suggest that both drugs have comparable efficacy in the R/R setting, with CR/CRh rates in the range of 23% to 26%.[10]
- Safety Profile Differentiation: This appears to be the most critical point of differentiation between the two agents. While Ziftomenib's key toxicity is Differentiation Syndrome, revumenib is associated with a significant and frequently reported risk of QTc prolongation, a cardiac safety concern that may require careful monitoring and could limit its use in certain patients.[23]
- Other Menin Inhibitors in Development: The clinical pipeline includes several other menin inhibitors, such as DS-1594, BMF-219, JNJ-75276617, and DSP-5336.[20] However, Ziftomenib and revumenib are the clear front-runners, being years ahead in development.
The competitive dynamics between the two lead assets are summarized in Table 6. This head-to-head comparison clarifies that while the race is close, the two companies have pursued slightly different initial regulatory strategies (NPM1-m for Ziftomenib vs. KMT2A-r for revumenib) and their products possess distinct safety profiles. This differentiation will likely be a key driver of market segmentation and physician choice. The ultimate commercial success for both agents, however, will likely be determined in the much larger frontline setting, where both companies are aggressively pursuing pivotal combination therapy trials. The exceptionally strong preliminary data from Ziftomenib's KOMET-007 trial has set a very high bar in this next competitive arena.
Table 6: Comparative Profile of Ziftomenib vs. Revumenib
| Feature | Ziftomenib (Kura Oncology / Kyowa Kirin) | Revumenib (Syndax Pharmaceuticals) | Source(s) |
|---|
| Lead Indication for NDA | R/R NPM1-mutant AML | R/R KMT2A-rearranged Acute Leukemia | 14 |
| Key Pivotal Trial | KOMET-001 (NCT04067336) | AUGMENT-101 (NCT04065399) | 9 |
| CR/CRh Rate (in lead indication) | 25% (NPM1-m AML) | 23% (KMT2A-r Leukemia) | 29 |
| Key Safety Signal | Differentiation Syndrome (DS) | QTc Prolongation | 10 |
| Development Status | NDA accepted with Priority Review; PDUFA date Nov 30, 2025 | NDA under review; potential approval late 2024 | 15 |
IX. Synthesis, Recommendations, and Future Outlook
Synthesis of Benefit-Risk Profile
Ziftomenib has established a highly favorable benefit-risk profile, particularly for its initial target population of adult patients with relapsed/refractory NPM1-mutant AML. In a setting characterized by a lack of targeted options and extremely poor outcomes, the ability of Ziftomenib monotherapy to induce deep, durable, and MRD-negative remissions represents a clinically meaningful benefit. This provides a crucial bridge to potentially curative stem cell transplantation for some patients and a significant extension of response for others. In the frontline setting, preliminary data suggest that its addition to standard chemotherapy could be transformative, yielding near-universal remission rates. The primary risk associated with Ziftomenib is Differentiation Syndrome, a serious but manageable on-target toxicity for which effective management protocols have been developed. The overall safety profile, notably the absence of a significant cardiac QTc prolongation signal, further strengthens its positive benefit-risk assessment.
Potential Place in Therapy
Based on the available evidence, the therapeutic role of Ziftomenib is expected to evolve rapidly following its potential approval.
- Initial Approval: Upon its anticipated approval, Ziftomenib is poised to become the standard of care for adult patients with relapsed or refractory NPM1-mutant AML. It will be the first and only targeted therapy approved for this specific, genetically defined patient population.
- Future Use: The compelling data from the KOMET-007 trial strongly support Ziftomenib's potential to be incorporated into the frontline standard of care for newly diagnosed patients with both NPM1-mutant and KMT2A-rearranged AML. Pending the successful completion of the Phase 3 KOMET-017 trials, Ziftomenib could become a foundational component of induction therapy for a large subset of AML patients. Further exploration in roles such as post-transplant maintenance therapy to prevent relapse is also a logical and promising avenue.
Future Research and Unanswered Questions
While the clinical program for Ziftomenib is robust, several important questions remain that will be the focus of future research.
- Mechanisms of Resistance: As with all targeted therapies, the emergence of resistance is inevitable. While a single acquired MEN1 resistance mutation has been identified in a patient progressing on Ziftomenib, the broader landscape of primary and acquired resistance mechanisms is not yet fully understood and requires systematic investigation.[32]
- Long-Term Outcomes: The ultimate measure of a cancer drug's success is its impact on survival. Mature overall survival data from both the KOMET-001 and KOMET-007 trials are needed to confirm that the high rates of deep remission translate into a durable long-term benefit for patients.
- Broader Patient Populations: The menin-KMT2A axis is also implicated in other leukemias, such as those with NUP98 rearrangements.[20] Investigating the efficacy of Ziftomenib in these and other potentially susceptible hematologic malignancies could further expand its clinical utility.
Final Conclusion
Ziftomenib stands as a landmark achievement in the development of precision medicines for Acute Myeloid Leukemia. Its highly specific mechanism of action, validated by strong clinical efficacy in genetically defined patient populations, has paved a clear regulatory path toward imminent approval. It is set to address a significant unmet need for patients with NPM1-mutant AML and holds the potential to redefine the standard of care for a much broader group of newly diagnosed patients in the near future. Its ongoing development and competitive positioning against other emerging menin inhibitors will be a key area to watch, but its foundational data have already established it as one of the most important new agents in the modern treatment of acute leukemia.
Works cited
- Ziftomenib | C33H42F3N9O2S2 | CID 138497449 - PubChem, accessed September 12, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Ziftomenib
- Ziftomenib | CAS# 2134675-36-6 | menin-MLL inhibitor - MedKoo Biosciences, accessed September 12, 2025, https://www.medkoo.com/products/48374
- Ziftomenib dosing, indications, interactions, adverse effects, and more - Medscape Reference, accessed September 12, 2025, https://reference.medscape.com/drug/ziftomenib-4000546
- ziftomenib | Ligand page - IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 12, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=11680
- Ziftomenib - Wikipedia, accessed September 12, 2025, https://en.wikipedia.org/wiki/Ziftomenib
- Kura Oncology and Kyowa Kirin Announce Submission of New Drug Application for Ziftomenib to FDA, accessed September 12, 2025, https://www.kyowakirin.com/media_center/news_releases/2025/pdf/e20250408_01.pdf
- NPM1m Acute Myeloid Leukemia - Syndax, accessed September 12, 2025, https://syndax.com/focus-area/npm1m-acute-myeloid-leukemia/
- Synergistic Strategies for KMT2A-Rearranged Leukemias: Beyond Menin Inhibitor - MDPI, accessed September 12, 2025, https://www.mdpi.com/2072-6694/16/23/4017
- FDA Grants Priority Review to Ziftomenib in NPM1-Mutant R/R AML - OncLive, accessed September 12, 2025, https://www.onclive.com/view/fda-grants-priority-review-to-ziftomenib-in-npm1-mutant-r-r-aml
- Ziftomenib in relapsed or refractory acute myeloid leukaemia ..., accessed September 12, 2025, https://pubmed.ncbi.nlm.nih.gov/39362248/
- Paper: Ziftomenib Combined with Intensive Induction (7+3) in Newly ..., accessed September 12, 2025, https://ash.confex.com/ash/2024/webprogram/Paper198218.html
- Kura Oncology and Kyowa Kirin Report Positive Updated Combination Data for Ziftomenib in Newly Diagnosed AML at 2025 European, accessed September 12, 2025, https://www.kyowakirin.com/media_center/news_releases/2025/pdf/e20250612_01.pdf
- Ziftomenib: What is it and is it FDA approved? - Drugs.com, accessed September 12, 2025, https://www.drugs.com/history/ziftomenib.html
- FDA Accepts Kura Oncology's NDA for Ziftomenib in NPM1-Mutant AML with Priority Review, accessed September 12, 2025, https://trial.medpath.com/news/fea12af2e0ad7262/fda-accepts-kura-oncology-s-nda-for-ziftomenib-in-npm1-mutant-aml-with-priority-review
- Kura Oncology and Kyowa Kirin Announce FDA Acceptance and Priority Review of New Drug Application for Ziftomenib in Adults with Relapsed or Refractory NPM1-Mutant AML, accessed September 12, 2025, https://ir.kyowakirin.com/en/news/news7835965998115940697/main/0/link/e20250602_01.pdf
- 2134675-36-6 | Ziftomenib - ChemScene, accessed September 12, 2025, https://www.chemscene.com/2134675-36-6.html
- Ziftomenib | Histone Methyltransferase - TargetMol, accessed September 12, 2025, https://www.targetmol.com/compound/ziftomenib
- Pipeline | Kura Oncology, accessed September 12, 2025, https://kuraoncology.com/pipeline/
- kuraoncology.com, accessed September 12, 2025, https://kuraoncology.com/pipeline/#:~:text=That's%20the%20mechanism%20underlying%20an,certain%20kinds%20of%20leukemia%20cells.
- Acute Myeloid Leukemia Research: How do menin-KMT2A inhibitors work? - CST Blog, accessed September 12, 2025, https://blog.cellsignal.com/menin-kmt2a-inhibitors-antibodies-for-studying-kmt2a-rearranged-aml
- Menin inhibition with revumenib for NPM1-mutated relapsed or refractory acute myeloid leukemia: the AUGMENT-101 study | Blood | American Society of Hematology, accessed September 12, 2025, https://ashpublications.org/blood/article/146/9/1065/537139/Menin-inhibition-with-revumenib-for-NPM1-mutated
- A 2024 Update on Menin Inhibitors. A New Class of Target Agents against KMT2A-Rearranged and NPM1-Mutated Acute Myeloid Leukemia - PubMed, accessed September 12, 2025, https://pubmed.ncbi.nlm.nih.gov/38651453/
- Drawing First Blood: Will Menin Inhibitors Revolutionize the ..., accessed September 12, 2025, https://ashpublications.org/ashclinicalnews/news/8059/Drawing-First-Blood-Will-Menin-Inhibitors
- Ziftomenib | CAS NO.:2134675-36-6 - GlpBio, accessed September 12, 2025, https://www.glpbio.com/ziftomenib.html
- Pharmacokinetics and ADME Characterization After Oral and ..., accessed September 12, 2025, https://pubmed.ncbi.nlm.nih.gov/39928533/
- Kura Oncology Presents Updated Clinical Data from KOMET-001 Trial of Menin Inhibitor Ziftomenib at ASH Annual Meeting | Blood Cancer United, accessed September 12, 2025, https://bloodcancerunited.org/resources/newsroom/kura-oncology-presents-updated-clinical-data-komet-001-trial-menin-inhibitor-ziftomenib
- FDA Approval Is Sought for Ziftomenib in NMP1-Mutant R/R AML - OncLive, accessed September 12, 2025, https://www.onclive.com/view/fda-approval-is-sought-for-ziftomenib-in-nmp1-mutant-r-r-aml
- Kura Oncology and Kyowa Kirin Announce Positive Ziftomenib Monotherapy Registrational Trial and Positive FDA Feedback for Upcoming Frontline Combination Trial Designs, accessed September 12, 2025, https://ir.kuraoncology.com/news-releases/news-release-details/kura-oncology-and-kyowa-kirin-announce-positive-ziftomenib
- Ziftomenib in relapsed/refractory (R/R) NPM1-mutant acute myeloid leukemia (AML): Phase 1b/2 clinical activity and safety results from the pivotal KOMET-001 study. - ASCO Publications, accessed September 12, 2025, https://ascopubs.org/doi/10.1200/JCO.2025.43.16_suppl.6506
- P504: UPDATED DATA FOR ZIFTOMENIB IN PATIENTS WITH ..., accessed September 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10429668/
- Evaluating Single-Agent Ziftomenib's Performance in NPM1+ R/R AML - CancerNetwork, accessed September 12, 2025, https://www.cancernetwork.com/view/evaluating-single-agent-ziftomenib-s-performance-in-npm1-r-r-aml
- Ziftomenib Induces Clinical Activity in NPM1-Mutated R/R AML - Targeted Oncology, accessed September 12, 2025, https://www.targetedonc.com/view/ziftomenib-induces-clinical-activity-in-npm1-mutated-r-r-aml
- Ziftomenib With 7+3 Shows Early Promise in Newly Diagnosed NPM1-Mutated/KMT2A-Rearranged AML - OncLive, accessed September 12, 2025, https://www.onclive.com/view/ziftomenib-with-7-3-shows-early-promise-in-newly-diagnosed-npm1-mutated-kmt2a-rearranged-aml
- EU/3/23/2881 - orphan designation for treatment of acute myeloid leukaemia | European Medicines Agency (EMA), accessed September 12, 2025, https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu-3-23-2881
- NPM1 Mutation in AML: Prognosis & Treatments - HealthTree for Acute Myeloid Leukemia, accessed September 12, 2025, https://healthtree.org/aml/community/articles/npm1-mutation-treatment-options
- Targeting and Monitoring Acute Myeloid Leukaemia with Nucleophosmin-1 (NPM1) Mutation - PMC - PubMed Central, accessed September 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9958584/
- Targeted therapy in NPM1-mutated AML: Knowns and unknowns - PMC - PubMed Central, accessed September 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9552319/
- Immunotherapeutic Potential of Mutated NPM1 for the Treatment of Acute Myeloid Leukemia, accessed September 12, 2025, https://www.mdpi.com/2072-6694/16/20/3443
- Entirely Venetoclax-Based Nonintensive Treatment Regimen for Newly Diagnosed NPM1-Mutated Acute Myeloid Leukemia in the ELN Favorable-Risk Group | Blood | American Society of Hematology, accessed September 12, 2025, https://ashpublications.org/blood/article/144/Supplement%201/6032/527677/Entirely-Venetoclax-Based-Nonintensive-Treatment
- KMT2A-Rearranged AML: Aligning Care, Guidelines, and Access, accessed September 12, 2025, https://www.ajmc.com/peer-exchange/kmt2a-rearranged-aml-aligning-care-guidelines-and-access
- Recent Developments and Evolving Therapeutic Strategies in KMT2A ‐Rearranged Acute Leukemia - PMC - PubMed Central, accessed September 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11491690/
- Post transplant maintenance therapy with newly FDA-approved revumenib for high-risk KMT2Ar acute leukemia | Blood Advances | American Society of Hematology, accessed September 12, 2025, https://ashpublications.org/bloodadvances/article/9/8/1988/535734/Post-transplant-maintenance-therapy-with-newly-FDA
- A 2024 Update on Menin Inhibitors. A New Class of Target Agents against KMT2A-Rearranged and NPM1-Mutated Acute Myeloid Leukemia - PMC, accessed September 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11036224/
- Menin Inhibitor Revumenib Yields Clinically Meaningful Responses in R/R NPM1-Mutated AML | ASH Clinical News | American Society of Hematology, accessed September 12, 2025, https://ashpublications.org/ashclinicalnews/news/8689/Menin-Inhibitor-Revumenib-Yields-Clinically
- Syndax Presents New Revuforj® (revumenib) Data in Relapsed/Refractory mNPM1 and NUP98r Acute Leukemia from AUGMENT-101 Trial at EHA 2025 - Investors, accessed September 12, 2025, https://ir.syndax.com/news-releases/news-release-details/syndax-presents-new-revuforjr-revumenib-data-relapsedrefractory
- Revumenib for NPM1-Mutant AML Succeeds in Pivotal Trial - Targeted Oncology, accessed September 12, 2025, https://www.targetedonc.com/view/revumenib-for-npm1-mutant-aml-succeeds-in-pivotal-trial
- Revumenib (SNDX-5613): a promising menin inhibitor for the management of relapsed and refractory acute myeloid leukaemia (AML) - PMC, accessed September 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11060303/
- Menin Inhibition With Revumenib for KMT2A-Rearranged Relapsed or Refractory Acute Leukemia (AUGMENT-101) - PubMed, accessed September 12, 2025, https://pubmed.ncbi.nlm.nih.gov/39121437/
- Menin inhibitor ziftomenib (KO-539) synergizes with drugs targeting chromatin regulation or apoptosis and sensitizes acute myeloid leukemia with MLL rearrangement or NPM1 mutation to venetoclax, accessed September 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10543165/