Small Molecule
C28H35Cl2NO5S
1352066-68-2
Navtemadlin is an investigational, orally bioavailable, small-molecule therapeutic representing a significant advancement in the targeted inhibition of the murine double minute 2 (MDM2) protein. Developed initially by Amgen as AMG-232 and now advanced by Kartos Therapeutics as KRT-232, Navtemadlin is engineered to be a potent and highly selective antagonist of the MDM2-p53 protein-protein interaction. Its mechanism of action is centered on the restoration of the tumor suppressor function of the p53 protein, a critical regulator of cellular homeostasis often referred to as the "guardian of the genome." In malignancies characterized by wild-type TP53 status but dysregulated by MDM2 overexpression, Navtemadlin competitively binds to MDM2, liberating p53 from negative regulation. This leads to p53 stabilization, accumulation, and subsequent transcriptional activation of target genes that orchestrate cell cycle arrest, senescence, and apoptosis in malignant cells.[1]
The clinical development of Navtemadlin has been most pronounced in myelofibrosis (MF), a severe hematologic malignancy with limited treatment options, particularly for patients who are relapsed or refractory (R/R) to Janus kinase (JAK) inhibitors. The landmark Phase III BOREAS trial (NCT03662126) demonstrated that Navtemadlin monotherapy achieved statistically significant and clinically meaningful improvements in key disease endpoints compared to the best available therapy (BAT). Specifically, Navtemadlin nearly tripled the rate of spleen volume reduction (SVR) of at least 35% and doubled the rate of total symptom score (TSS) improvement of at least 50% at 24 weeks.[4]
Beyond symptomatic improvement, Navtemadlin has shown compelling evidence of disease-modifying activity, a critical unmet need in MF. Treatment has been associated with significant reductions in bone marrow fibrosis, circulating CD34+ malignant progenitor cells, and the variant allele frequency (VAF) of driver gene mutations.[7] These findings suggest that Navtemadlin targets the underlying clonal cell population responsible for the disease. The safety profile of Navtemadlin is well-characterized, predictable, and considered manageable. The most common adverse events are on-target effects related to p53 activation in rapidly dividing normal tissues, primarily manifesting as reversible gastrointestinal toxicities (nausea, diarrhea) and hematologic cytopenias (thrombocytopenia, neutropenia).[4] The implementation of an intermittent dosing schedule has been pivotal in establishing a therapeutic window that balances robust efficacy with tolerability.
Having received Orphan Drug and Fast Track designations from regulatory agencies, Navtemadlin is poised to become a first-in-class approved therapy for R/R myelofibrosis. Ongoing and planned clinical trials are exploring its utility in combination with ruxolitinib in earlier lines of MF therapy and its potential in other TP53-wild-type malignancies, positioning Navtemadlin as a transformative agent in oncology.
This section provides a definitive summary of Navtemadlin's nomenclature, standard identifiers, and fundamental chemical and physical properties that govern its formulation, stability, and biological activity.
Navtemadlin is identified by a consistent set of names and codes across scientific literature and regulatory databases, ensuring unambiguous reference.
Navtemadlin is a synthetic small molecule designed through a structure-based approach to optimize its interaction with the MDM2 protein.
The physicochemical profile of Navtemadlin has profound implications for its development and clinical application. The combination of very low aqueous solubility and high lipophilicity is a classic hallmark of a Biopharmaceutics Classification System (BCS) Class II compound. For such molecules, oral absorption is typically limited not by their ability to cross cell membranes (permeability) but by the rate at which they can dissolve in the gastrointestinal fluids. This dissolution-rate-limited absorption often leads to pharmacokinetic variability and susceptibility to food effects, where the presence of fats and bile salts in the gut can enhance solubilization and increase drug exposure. This fundamental chemical nature directly informed the clinical development path, making it necessary to conduct a dedicated clinical study [20] to precisely characterize the impact of food and formulation changes on Navtemadlin's pharmacokinetic profile, ensuring consistent and predictable dosing for patients.[20]
The therapeutic activity of Navtemadlin is rooted in its precise modulation of the p53-MDM2 signaling axis, a central pathway in cancer biology. Its exceptional potency and selectivity for its molecular target are the foundation of both its efficacy and its characteristic safety profile.
The tumor suppressor protein p53 is a transcription factor that plays a pivotal role in maintaining genomic integrity. In response to cellular stress, such as DNA damage or oncogene activation, p53 is activated and orchestrates a range of cellular responses, including transient cell cycle arrest to allow for DNA repair, or permanent arrest (senescence) or programmed cell death (apoptosis) to eliminate irreparably damaged cells.[2] Due to this critical function, the
TP53 gene is the most frequently mutated gene in human cancers.
However, in a substantial subset of cancers, the p53 protein itself is wild-type (TP53-WT) but its function is abrogated by other mechanisms. The primary negative regulator of p53 is the murine double minute 2 (MDM2) protein.[3] MDM2 is an E3 ubiquitin-protein ligase that physically binds to p53, performing two key inhibitory functions: first, it targets p53 for ubiquitination and subsequent degradation by the proteasome, thereby keeping cellular p53 levels low; second, it binds to the transactivation domain of p53, directly blocking its ability to activate the transcription of its target genes.[2] In many
TP53-WT tumors, such as certain sarcomas, leukemias, and myelofibrosis, the MDM2 gene is amplified or the protein is overexpressed, effectively silencing p53 function and allowing cancer cells to proliferate unchecked.[3] This creates a clear therapeutic vulnerability: inhibiting the MDM2-p53 interaction in these tumors can reactivate the endogenous p53 pathway, representing a powerful anti-cancer strategy.[2]
Navtemadlin was developed through structure-based drug design to be a potent and competitive inhibitor of the MDM2-p53 protein-protein interaction (PPI).[2] It functions by occupying the deep hydrophobic cleft on the surface of the MDM2 protein that is the natural binding site for the p53 transactivation domain. By physically blocking this site, Navtemadlin prevents MDM2 from binding to and inhibiting p53.[13]
The potency of this interaction is exceptionally high, as quantified by multiple biophysical and biochemical assays:
A critical feature of Navtemadlin is its selectivity. Affinity-based protein profiling experiments using photoactivatable probes have robustly confirmed that MDM2 is the primary and dominant molecular target in cancer cells, with minimal engagement of other proteins.[1] The drug shows no significant activity against other MDM family members at concentrations up to 10 µM and has negligible off-target activity against a large panel of kinases, underscoring its highly targeted nature.[26]
By effectively uncoupling p53 from MDM2-mediated degradation and inhibition, Navtemadlin triggers a cascade of downstream events consistent with p53 pathway activation.[2] The immediate consequence is the rapid stabilization and accumulation of p53 protein within the tumor cell. This activated p53 then functions as a transcription factor, binding to the promoter regions of its target genes and upregulating their expression.[25]
Key p53 target genes induced by Navtemadlin include:
The integrated cellular outcomes of this transcriptional program are profound: tumor cells undergo cell cycle arrest and apoptosis, leading to a potent inhibition of proliferation and, in preclinical models, durable tumor regression.[2] This anti-tumor activity is strictly dependent on a functional, wild-type p53 protein. In cell lines where
TP53 is mutated or deleted, Navtemadlin shows no significant effect on cell growth, confirming its on-target mechanism of action.[13]
The activation of the p53 pathway by Navtemadlin leads to measurable changes in downstream biomarkers that can be monitored in patients to confirm target engagement and pharmacodynamic activity. The most well-validated of these is Macrophage Inhibitory Cytokine-1 (MIC-1), also known as Growth Differentiation Factor-15 (GDF15). MIC-1 is a direct transcriptional target of p53. Clinical studies have consistently shown that administration of Navtemadlin leads to a robust, dose- and plasma concentration-dependent increase in serum MIC-1 levels.[20] The time course of MIC-1 induction, with a peak delayed by approximately 8 hours relative to the drug's peak concentration, is consistent with a process involving gene transcription and protein synthesis.[27] This makes MIC-1 an invaluable clinical pharmacodynamic biomarker for verifying that Navtemadlin is effectively engaging MDM2 and activating the p53 pathway in patients.[20]
The highly selective and potent on-target mechanism of Navtemadlin is the cornerstone of its therapeutic potential. However, this specificity is also inextricably linked to its toxicity profile. The p53 protein is a universal regulator of cell fate, and its potent activation is not confined to cancer cells. Rapidly proliferating normal tissues, most notably the hematopoietic progenitors in the bone marrow and the epithelial lining of the gastrointestinal tract, are also highly sensitive to p53-mediated cell cycle arrest and apoptosis. Consequently, the potent reactivation of p53 by Navtemadlin in these tissues is the direct cause of the most common and dose-limiting toxicities observed in clinical trials: hematologic cytopenias (thrombocytopenia, neutropenia) and gastrointestinal disturbances (nausea, vomiting, diarrhea). These adverse events are not off-target effects but rather an expected pharmacologic consequence of the drug's intended mechanism, a factor that has profoundly influenced the development of dosing schedules designed to manage these on-target liabilities.[4]
The clinical utility of Navtemadlin is underpinned by a pharmacokinetic (PK) profile that allows for convenient oral administration and sustained target engagement. Its absorption, distribution, metabolism, and excretion (ADME) properties have been characterized in preclinical species and extensively in humans across multiple clinical trials.
Navtemadlin exhibits predictable PK properties that support a once-daily, intermittent dosing schedule.
Navtemadlin's pharmacokinetics are predictable across a wide range of doses. Clinical studies have demonstrated that exposure, as measured by AUC, increases in a dose-proportional manner over a dose range of 15 mg to 480 mg, indicating linear pharmacokinetics.[20]
Population PK modeling, which analyzes data from a large and diverse patient population, has identified certain patient characteristics, or covariates, that can influence drug exposure. A significant finding is that patients with acute myeloid leukemia (AML) exhibit a 61.6% greater steady-state AUC compared to patients with solid tumors, suggesting potential differences in drug absorption or clearance between these populations.[30] Furthermore, a correlation was found with serum albumin levels; patients with decreased albumin (e.g., at the 5th percentile of the population) were modeled to have a 47.7% increase in AUC, likely due to reduced plasma protein binding and a higher fraction of unbound, clearable drug.[30]
The pharmacokinetic profile of Navtemadlin reveals a critical dichotomy between its systemic efficacy and its potential for treating CNS malignancies. While its systemic exposure is robust, its ability to cross the blood-brain barrier (BBB) is severely restricted. Preclinical studies in mice have unequivocally demonstrated that Navtemadlin is a substrate for active efflux transporters at the BBB, particularly P-glycoprotein (P-gp, encoded by the ABCB1 gene).[21] This active pumping of the drug out of the brain results in an extremely low brain-to-plasma concentration ratio (
Kp,brain) of just 0.009.[21]
The functional consequence of this poor CNS penetration is stark. In orthotopic mouse models of glioblastoma (GBM), where the tumor is grown within the brain, Navtemadlin was completely ineffective even at high doses (100 mg/kg).[21] However, a pivotal experiment using genetically engineered mice lacking the key efflux pumps (
Abcb1a/b and Abcg2) provided definitive proof-of-principle. In these efflux-deficient mice, a much lower dose of Navtemadlin (25 mg/kg) was able to achieve therapeutic concentrations in the brain tumor and significantly extended survival.[21] This demonstrates that the drug is inherently active against GBM cells, but its efficacy is nullified by the delivery barrier.
These preclinical findings were directly translated and confirmed in a Phase 0 "window-of-opportunity" study in human GBM patients. This type of study involves administering the drug to patients shortly before a scheduled surgery, allowing for direct measurement of drug concentrations in the resected tumor tissue. The results showed that at the clinically relevant 240 mg dose, the minimum effective tumor exposures (as determined from preclinical models) were achieved in only a small minority of patients (3 out of 16).[21] This confirmation of inadequate drug delivery to the CNS explains why the clinical development of Navtemadlin for primary brain tumors has not progressed, while its development for systemic diseases like myelofibrosis and other hematologic cancers, where the BBB is not a factor, has advanced successfully.[14] This illustrates how a single pharmacokinetic property can fundamentally dictate a drug's entire clinical development strategy.
The clinical investigation of Navtemadlin has been extensive, evolving from broad, early-phase exploration across numerous cancer types to a highly focused, late-stage program targeting indications with the strongest biological rationale and clinical signal. This strategic evolution has positioned Navtemadlin for potential regulatory approval in myelofibrosis.
Navtemadlin has been evaluated as a monotherapy and in combination with other anti-cancer agents in multiple Phase I, II, and III clinical trials. The breadth of this program reflects the foundational importance of the p53-MDM2 pathway across oncology. Table 1 provides a summary of the key clinical trials that have defined its development trajectory.
Table 1: Summary of Key Navtemadlin Clinical Trials
NCT Identifier | Trial Name/Acronym | Phase | Indication(s) | Status (as of latest data) | Key Design/Objectives |
---|---|---|---|---|---|
NCT03662126 | BOREAS | 2/3 | Relapsed/Refractory Myelofibrosis (R/R MF) | Active, Not Recruiting | Randomized, open-label study of Navtemadlin vs. Best Available Therapy (BAT) 3 |
NCT06479135 | POIESIS | 3 | JAK Inhibitor-Naïve Myelofibrosis (with suboptimal response) | Recruiting | Randomized, double-blind, add-on study of Navtemadlin vs. Placebo with Ruxolitinib 39 |
NCT04485260 | KRT-232-109 | 1b/2 | Myelofibrosis (with suboptimal response to Ruxolitinib) | Active, Not Recruiting | Open-label study of Navtemadlin added to stable-dose Ruxolitinib 40 |
NCT03787602 | KRT-232-103 | 1b/2 | Merkel Cell Carcinoma (MCC) (post-PD-1/L1) | Active, Not Recruiting | Dose-finding and expansion study of Navtemadlin monotherapy 42 |
NCT03041688 | NCI-2017-00152 | 1b | Acute Myeloid Leukemia (AML) (R/R or Newly Diagnosed) | Active, Not Recruiting | Dose-escalation study of Navtemadlin + Decitabine + Venetoclax 10 |
NCT02016729 | - | 1b | Acute Myeloid Leukemia (AML) | Completed | Study of Navtemadlin alone and in combination with Trametinib 44 |
NCT01723020 | - | 1 | Advanced Solid Tumors, Multiple Myeloma | Completed | First-in-human dose-escalation and expansion study 45 |
NCT04113616 | KRT-232-104 | 1b/2 | AML secondary to MPN | Active, Not Recruiting | Open-label study evaluating different Navtemadlin dosing schedules 46 |
NCT03220339 | ALLIANCE-ABTC-1604 | 0 | Recurrent Glioblastoma (GBM) | Active, Not Recruiting | Window-of-opportunity study to assess CNS penetration 47 |
The strongest efficacy signals for Navtemadlin have emerged in hematologic malignancies, particularly myeloproliferative neoplasms (MPNs).
Myelofibrosis has become the lead indication for Navtemadlin, driven by a strong biological rationale (MDM2 is overexpressed in malignant MF CD34+ cells) and compelling clinical data.[3]
Early-phase studies have explored Navtemadlin in AML, where TP53-WT status is common. A Phase Ib study (NCT02016729) evaluated Navtemadlin alone and in combination with the MEK inhibitor trametinib, establishing safety and dose-proportional PK, with some patients achieving stable disease.[33] A subsequent Phase Ib trial (NCT03041688) is investigating a triplet combination of Navtemadlin with decitabine and the BCL-2 inhibitor venetoclax, a modern standard-of-care backbone in AML.[10]
The clinical program has also included a Phase II study in phlebotomy-dependent polycythemia vera (PV) (NCT03669965) [52] and an ongoing Phase Ib/II study in relapsed/refractory Philadelphia chromosome-positive chronic myeloid leukemia (CML) in combination with a tyrosine kinase inhibitor (TKI) (NCT04835584).[53]
The clinical development path of Navtemadlin exemplifies a successful pharmaceutical strategy. An initial broad exploration by the originator, Amgen, across a wide array of cancers identified a particularly strong signal in myelofibrosis. The current developer, Kartos Therapeutics, has astutely capitalized on this by executing a focused and rigorous late-stage program in MF. This strategy of "finding the right drug for the right disease" has propelled Navtemadlin to the cusp of approval and established a clear path for building a franchise around this novel agent, starting with the highest unmet need in the R/R setting and systematically moving into earlier lines of therapy.
While the primary focus has shifted to hematologic cancers, Navtemadlin has also been evaluated in several solid tumor types, with notable activity seen in Merkel cell carcinoma.
MCC is a rare but aggressive neuroendocrine skin cancer. A Phase Ib/II study (NCT03787602) was designed to evaluate Navtemadlin in patients with TP53-WT MCC whose disease had progressed after treatment with anti-PD-1/L1 immunotherapy.[42] The trial identified a recommended Phase 2 dose and schedule of 180 mg daily for 5 days, followed by 23 days off. In this heavily pretreated population, Navtemadlin monotherapy demonstrated promising and durable anti-tumor activity, achieving a confirmed objective response rate (ORR) of 25% and a disease control rate of 63%. Some responses were remarkably durable, with one patient achieving a complete metabolic remission after two years on treatment, establishing proof-of-concept for MDM2 inhibition in this disease.[42]
As detailed in the Pharmacokinetics section, the development of Navtemadlin for GBM was halted by a fundamental drug delivery challenge. A Phase 0 window-of-opportunity study confirmed that, due to active efflux by P-glycoprotein at the blood-brain barrier, therapeutically relevant concentrations of Navtemadlin could not be achieved in the majority of patients' brain tumors.[21] This finding, while disappointing, provided a clear, data-driven rationale to discontinue development in this indication and focus resources on systemic cancers where the drug could reliably reach its target.
Navtemadlin's clinical journey began with a first-in-human Phase I study (NCT01723020) that included patients with a variety of advanced solid tumors and multiple myeloma, which successfully established the drug's safety profile and recommended dose.[13] Other explorations have included a trial in soft tissue sarcoma combined with radiation therapy [54] and an ongoing Phase II study in
TP53-WT relapsed/refractory small cell lung cancer (SCLC).[55]
The safety profile of Navtemadlin has been extensively characterized across numerous clinical trials involving hundreds of patients. The observed adverse events are largely predictable, manageable, and consistent with the drug's on-target mechanism of p53 activation in rapidly dividing normal tissues.
Navtemadlin has demonstrated an acceptable and manageable safety profile, with the majority of treatment-emergent adverse events (TEAEs) being Grade 1 or 2 in severity and reversible upon cessation of dosing.[4] The most frequently reported and clinically significant toxicities fall into two main categories: gastrointestinal and hematologic. The combination of Navtemadlin with ruxolitinib in myelofibrosis patients appears to be well-tolerated, with a safety profile generally consistent with that of each agent alone.[9] Table 2 summarizes the incidence of common TEAEs from key myelofibrosis trials.
Table 2: Incidence of Common Treatment-Emergent Adverse Events (TEAEs) in Myelofibrosis Trials
Adverse Event | Navtemadlin Monotherapy (BOREAS) 4 | Navtemadlin + Ruxolitinib (NCT04485260) 9 |
---|---|---|
Any Grade (%) | Grade ≥3 (%) | |
Gastrointestinal | ||
Nausea | 42 | 4 |
Diarrhea | 41 | 6 |
Vomiting | 25 | 2 |
Hematologic | ||
Thrombocytopenia | 46 | 37 |
Anemia | 36 | 29 |
Neutropenia | 30 | 25 |
Constitutional | ||
Fatigue/Asthenia | N/A | N/A |
The on-target nature of Navtemadlin's adverse events allows for proactive monitoring and management.
In the initial Phase I dose-escalation studies, dose-limiting toxicities (DLTs) were encountered at higher dose levels. Specifically, Grade 3/4 thrombocytopenia and neutropenia were identified as DLTs at doses of 360 mg and 480 mg administered daily for 7 days.[13] These findings were instrumental in guiding dose selection for later-phase studies. The 240 mg daily dose for 7 days in a 28-day cycle was selected as the Recommended Phase 2 Dose (RP2D) for the pivotal BOREAS study, as it was determined to provide a favorable balance between robust anti-tumor efficacy and a manageable safety profile.[13]
The successful clinical application of Navtemadlin hinges on its dosing schedule. The potent, on-target mechanism would likely lead to unacceptable cumulative toxicity if administered continuously. The intermittent schedule (e.g., 7 days on, 21 days off) is a critical design feature that creates the therapeutic window. The 7-day "on" period is sufficient to exert a powerful apoptotic effect on malignant cells, as evidenced by the efficacy data. The subsequent 21-day "off" period allows the normal, healthy bone marrow and gastrointestinal tissues to recover from the transient p53-induced stress. This "pulsed" therapeutic approach is a sophisticated application of pharmacodynamic principles that effectively manages the on-target liabilities of the drug, enabling its long-term administration and unlocking its clinical potential.
Navtemadlin is an investigational agent that has not yet received marketing approval from any regulatory agency. However, its promising clinical data in areas of high unmet medical need have earned it several special designations that are intended to facilitate and expedite its development and review.
Navtemadlin's potential has been recognized by key global health authorities:
These designations provide benefits such as protocol assistance, reduced fees, and potential market exclusivity upon approval, highlighting the regulatory support for Navtemadlin's development.
The therapeutic strategy of inhibiting the MDM2-p53 interaction has been pursued for over two decades, but the clinical development of MDM2 inhibitors has been challenging. Early candidates like Nutlin-3a had poor pharmacokinetic properties, and other clinical-stage molecules, such as Idasanutlin (RG7388), have faced setbacks in pivotal trials.[58] The field has been hampered by a narrow therapeutic window, with on-target hematologic and GI toxicities often limiting the ability to achieve durable anti-tumor responses with monotherapy.[58] In this context, the positive results of the Phase III BOREAS trial are a landmark achievement, not only for Navtemadlin but for the entire class of MDM2 inhibitors. It provides the first robust, large-scale validation of this mechanism as a viable and effective single-agent therapy in a well-defined patient population.
The future direction for Navtemadlin is clearly defined. The immediate goal is to secure regulatory approval for the treatment of relapsed/refractory myelofibrosis based on the BOREAS data, which would establish it as the first non-JAK inhibitor therapy to demonstrate significant efficacy in this setting. The long-term strategy involves expanding its role within the MF treatment paradigm. The ongoing POIESIS study is a critical step in this direction, evaluating if adding Navtemadlin to ruxolitinib in the first-line setting for patients with a suboptimal response can improve outcomes and potentially change the standard of care.[6] Continued development in other
TP53-WT malignancies with a strong biological rationale, such as Merkel cell carcinoma and small cell lung cancer, also remains a key area of interest.
In conclusion, Navtemadlin has progressed from a highly potent molecular entity to a late-stage clinical asset with proven efficacy and manageable safety in a disease with dire need for new therapies. Its success is a testament to the power of targeted drug development, the importance of understanding on-target toxicity, and the strategic implementation of dosing schedules and clinical trial designs to create a viable therapeutic window. Pending regulatory approval, Navtemadlin is poised to offer a novel, disease-modifying treatment option for patients with myelofibrosis and represents a significant validation of the MDM2-p53 axis as a druggable target in oncology.
Published at: September 7, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.