MedPath

Azenosertib Advanced Drug Monograph

Published:Sep 19, 2025

Generic Name

Azenosertib

Drug Type

Small Molecule

Chemical Formula

C29H34N8O2

CAS Number

2376146-48-2

A Comprehensive Report on Azenosertib (ZN-c3): A First-in-Class WEE1 Inhibitor for Oncologic Indications

Executive Summary

Azenosertib (also known as ZN-c3) is an investigational, orally bioavailable, small molecule drug being developed by Zentalis Pharmaceuticals as a potentially first-in-class and best-in-class inhibitor of WEE1 kinase. The primary mechanism of action involves the targeted inhibition of WEE1, a critical regulator of the G2/M cell cycle checkpoint. This inhibition abrogates the cell's ability to arrest and repair DNA damage before mitosis, leading to a cascade of events including increased replication stress, premature mitotic entry, and ultimately, cancer cell death via mitotic catastrophe. This therapeutic strategy is particularly effective in tumors characterized by high genomic instability, such as those with p53 mutations or, most notably, overexpression of Cyclin E1, which has emerged as a key predictive biomarker for Azenosertib sensitivity.

The clinical development program for Azenosertib is extensive, evaluating the agent as both a monotherapy and in combination with chemotherapy, PARP inhibitors, and other molecularly targeted agents across a range of solid tumors. The program's strategy has been significantly refined by pharmacokinetic studies that established an optimized intermittent dosing schedule (400 mg daily, 5 days on, 2 days off), which more than doubles drug exposure while improving tolerability compared to continuous dosing.

Pivotal clinical data has demonstrated compelling anti-tumor activity, particularly in heavily pretreated gynecologic malignancies. In the Phase 2 DENALI trial, Azenosertib monotherapy achieved a confirmed objective response rate (ORR) of 34.9% in response-evaluable patients with Cyclin E1-positive, platinum-resistant ovarian cancer. Similarly, promising efficacy signals have been observed in uterine serous carcinoma. The safety profile is characterized primarily by manageable hematologic and gastrointestinal adverse events.

The regulatory path for Azenosertib is supported by a Fast Track Designation from the U.S. Food and Drug Administration (FDA) for its use in Cyclin E1-positive ovarian cancer. While the program faced a temporary partial clinical hold in 2024 due to two patient deaths, the hold was lifted without required changes to the clinical development plan, suggesting regulatory confidence in the drug's manageable risk-benefit profile. With no currently approved WEE1 inhibitors, Azenosertib is positioned as a leading candidate to address a significant unmet medical need in biomarker-defined patient populations with difficult-to-treat cancers.

Section 1: Introduction and Therapeutic Landscape

1.1. Azenosertib: A Novel Investigational Agent in Oncology

Azenosertib, also known by its developmental code ZN-c3, is a clinical-stage, small molecule therapeutic candidate representing a new frontier in precision oncology.[1] Developed by Zentalis Pharmaceuticals, it is engineered as a potent, selective, and orally bioavailable inhibitor of WEE1 G2 checkpoint kinase (WEE1).[2] The agent is being strategically advanced for the treatment of various advanced solid tumors and hematological malignancies, with a particular focus on cancers where existing therapies have limited efficacy.[4] Azenosertib is positioned as a potentially first-in-class therapeutic, as there are currently no FDA-approved drugs targeting the WEE1 kinase, and has been designed with the goal of achieving a best-in-class profile through superior selectivity and optimized pharmacokinetic properties.[1]

1.2. The Unmet Need in Cancers with High Genomic Instability

A foundational principle of cancer biology is the accumulation of genetic mutations and the resulting genomic instability that drives tumor development and progression.[3] Many of the most aggressive and difficult-to-treat cancers, such as high-grade serous ovarian carcinoma (HGSOC) and uterine serous carcinoma (USC), are defined by this characteristic.[1] A common feature of these genomically unstable tumors is the loss of function of key tumor suppressor genes, most notably

TP53.[7]

The p53 protein is a primary guardian of the genome, and one of its critical functions is to regulate the G1/S cell cycle checkpoint. This checkpoint serves as a crucial decision point, halting the cell cycle to allow for the repair of DNA damage before it is replicated in the S phase. When p53 is mutated or lost, this G1 checkpoint is often abrogated.[7] Consequently, these cancer cells become critically dependent on the remaining G2/M checkpoint to pause the cell cycle and repair DNA damage accumulated during replication, thereby preventing lethal errors during mitosis. This dependency creates a specific vulnerability, as the G2/M checkpoint becomes the sole gatekeeper of genomic integrity for the cancer cell—a vulnerability that is not present in normal, healthy cells with intact G1 checkpoints.[7]

1.3. Overview of WEE1 Kinase as a Therapeutic Target

The G2/M checkpoint is masterfully controlled by the WEE1 kinase, a serine/threonine-protein kinase that functions as a pivotal negative regulator of cell cycle progression.[2] The primary function of WEE1 is to phosphorylate and thereby inactivate Cyclin-Dependent Kinase 1 (CDK1, also known as Cdc2) and Cyclin-Dependent Kinase 2 (CDK2).[1] This inhibitory phosphorylation prevents the activation of the CDK1/Cyclin B complex, which is the master switch that triggers a cell's entry into mitosis.[7] By holding CDK1 in an inactive state, WEE1 effectively enforces the G2/M checkpoint, providing a critical window for the DNA Damage Response (DDR) network to repair any genetic lesions before cell division proceeds.[1]

The reliance of p53-deficient cancer cells on this single checkpoint for survival forms the basis of a therapeutic strategy known as synthetic lethality. In this context, the loss of p53 function is not lethal on its own, and the inhibition of WEE1 is generally tolerated by normal cells that can rely on their G1 checkpoint. However, in a cancer cell lacking the G1 checkpoint, the pharmacological inhibition of WEE1 becomes synthetically lethal. By removing the final stop sign, WEE1 inhibition forces the cancer cell to proceed into mitosis with a high burden of unrepaired DNA damage, leading to catastrophic cell division and death.[7] Therefore, targeting WEE1 with an inhibitor like Azenosertib is a highly rational approach to selectively kill cancer cells while sparing healthy tissue.

Section 2: Molecular Profile and Physicochemical Characteristics

2.1. Chemical Identity and Structure

Azenosertib is a small molecule drug with a precisely defined chemical structure and identity.[1] Its International Nonproprietary Name (INN), Azenosertib, was formally proposed in 2022, while it is most frequently referred to in scientific literature and clinical development by its code, ZN-c3.[2] The molecule's structure is complex, reflected in its systematic IUPAC name: (R)-2-allyl-1-(7-ethyl-7-hydroxy-6,7-dihydro-5H-cyclopenta[b]pyridin-2-yl)-6-((4-(4-methylpiperazin-1-yl)phenyl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one.[8] This name describes a core pyrazolo[3,4-d]pyrimidine structure with several key functional substitutions that are responsible for its potent and selective binding to the WEE1 kinase.

2.2. Physicochemical and Formulation Properties

The molecular formula of Azenosertib is C29​H34​N8​O2​, corresponding to a molecular weight of approximately 526.63 g/mol.[8] As an orally active agent, its physicochemical properties have been optimized for bioavailability.[3] For research and preclinical use, Azenosertib is typically supplied as a light yellow to yellow solid powder.[9] It demonstrates good solubility in organic solvents such as dimethyl sulfoxide (DMSO).[11] For in vivo animal studies, specific formulation protocols have been developed to achieve adequate solubility and exposure, for instance, using a vehicle of 10% DMSO, 40% PEG300, 5% Tween-80, and 45% saline.[11] The compound is stable under standard storage conditions, with long-term storage of the powder recommended at -20°C.[8]

The following table provides a consolidated summary of Azenosertib's key identifiers and properties.

Table 1: Summary of Azenosertib's Chemical and Physical Properties

PropertyValueSource(s)
Official NameAzenosertib2
Developmental CodeZN-c38
DrugBank IDDB18028
CAS Number2376146-48-28
Drug TypeSmall Molecule1
Molecular FormulaC29​H34​N8​O2​8
Molecular Weight526.63 g/mol10
IUPAC Name(R)-2-allyl-1-(7-ethyl-7-hydroxy-6,7-dihydro-5H-cyclopenta[b]pyridin-2-yl)-6-((4-(4-methylpiperazin-1-yl)phenyl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one8
InChIKeyOXTSYWDBUVRXFF-GDLZYMKVSA-N2
SMILES CodeO=C1N(CC=C)N(C2=CC=C(CC[C@@]3(CC)O)C3=N2)C4=NC(NC5=CC=C(N6CCN(C)CC6)C=C5)=NC=C418

Section 3: In-Depth Pharmacology and Mechanism of Action

3.1. The WEE1 Pathway in Cell Cycle Control and DNA Damage Repair

The WEE1 kinase is an indispensable component of the cellular machinery that maintains genomic integrity. It functions as the primary gatekeeper of the G2/M transition, one of the most critical checkpoints in the cell division cycle.[1] In response to DNA damage detected during the S or G2 phases, the DDR network activates WEE1. The activated WEE1 kinase then carries out its inhibitory function by phosphorylating specific tyrosine residues on its key substrates, CDK1 and CDK2.[1] This phosphorylation event, particularly at the Tyr15 residue of CDK1, prevents the formation of an active CDK1/Cyclin B complex, which is the master engine that drives the cell into mitosis.[3] By effectively applying this brake, WEE1 enforces a cell cycle arrest, providing the cell with the necessary time to recruit repair machinery and correct any DNA lesions before the high-risk process of chromosome segregation begins.[1]

3.2. Azenosertib's Potency, Selectivity, and Target Engagement

Azenosertib has demonstrated highly potent and specific inhibition of the WEE1 kinase. In biochemical assays, it exhibits a half-maximal inhibitory concentration (IC50​) of approximately 3.9 nM, indicating very strong binding to its target.[10] This potency is a critical attribute for achieving a therapeutic effect at clinically manageable doses.

Beyond potency, a key feature that supports Azenosertib's potential best-in-class profile is its high degree of selectivity.[1] In a broad kinase panel screen against 476 kinases, Azenosertib showed significant inhibition of only five, with WEE1 being the primary target.[6] A particularly important point of differentiation is its selectivity over Polo-like kinase 1 (PLK1), another key mitotic regulator. Off-target inhibition of PLK1 has been associated with significant toxicities, including neutropenia and bone marrow suppression.[6] Azenosertib is approximately 60-fold more potent against WEE1 (

IC50​ = 3.8 nM) than against PLK1 (IC50​ = 227 nM). Furthermore, cellular assays confirmed this selectivity, showing no substantial functional inhibition of PLK1 activity at concentrations where WEE1 is fully inhibited.[6] This superior selectivity profile is designed to minimize off-target toxicities and widen the therapeutic window of the drug.

Evidence of direct target engagement within cancer cells has been robustly demonstrated. Treatment with Azenosertib leads to a marked reduction in the levels of phosphorylated CDK1 at tyrosine 15 (pY15-CDK1), the direct downstream substrate of WEE1. This reduction serves as a pharmacodynamic biomarker, confirming that the drug is effectively inhibiting its intended target in a cellular context.[3]

3.3. Cellular Consequences of WEE1 Inhibition: Inducing Mitotic Catastrophe

The pharmacological inhibition of WEE1 by Azenosertib sets off a lethal chain of events in vulnerable cancer cells. First, it abrogates the G2/M checkpoint, forcing cells to enter mitosis prematurely, even in the presence of significant, unrepaired DNA damage.[1] Second, Azenosertib exacerbates the underlying genomic instability by inducing further replication stress. This is achieved by causing aberrant firing of DNA replication origins and depleting the cellular pools of nucleotides required for DNA synthesis, leading to an even greater burden of DNA damage.[1]

The convergence of these two effects—forcing division while simultaneously increasing the damage to be divided—is catastrophic for the cell. As the cell attempts to segregate its damaged and broken chromosomes during mitosis, it fails, leading to a terminal cellular state known as mitotic catastrophe. This process ultimately triggers apoptosis, or programmed cell death, resulting in the elimination of the cancer cell.[1] This multi-pronged mechanism of action—disrupting checkpoints, increasing DNA damage, and forcing lethal cell division—underpins the potent antitumor activity of Azenosertib.

Section 4: Preclinical Validation and Clinical Rationale

4.1. Evidence of Antitumor Activity in Preclinical Models

The therapeutic hypothesis for Azenosertib has been extensively validated in preclinical settings, where it has demonstrated robust and broad antitumor activity. In numerous cancer cell line and xenograft models, Azenosertib has shown potent inhibition of tumor growth.[6] For instance, in a xenograft model using A427 lung cancer cells, daily oral administration of Azenosertib at 80 mg/kg for 28 days resulted in significant tumor growth inhibition.[9] A notable finding from these studies is the durability of the response; Azenosertib has been shown to be highly effective at delaying the duration of tumor regrowth even after the cessation of treatment, suggesting that it induces a profound and lasting cytotoxic effect rather than a temporary cytostatic one.[6] This broad preclinical activity provided a strong foundation for its advancement into clinical trials across multiple solid tumor indications.

4.2. Pharmacokinetic (PK) Profile and Dosing Regimen Optimization

Azenosertib was rationally designed to possess superior pharmacokinetic (PK) properties suitable for an oral anti-cancer agent.[1] Preclinical studies confirmed these favorable characteristics. In a beagle dog model, a 10 mg/kg oral dose yielded a maximum plasma concentration (

Cmax​) of 2.1 μM, a plasma half-life (T1/2​) of 2.3 hours, and an exceptional oral bioavailability (F) of 142%, indicating efficient absorption and systemic exposure.[9]

In the clinical setting, the optimization of the dosing regimen proved to be a pivotal step in unlocking the drug's full therapeutic potential. Initial studies explored a continuous daily dosing schedule. However, data from the Phase 1 ZN-c3-001 dose optimization trial revealed that an intermittent dosing schedule was superior.[16] The company established a recommended Phase 2 dose (RP2D) for monotherapy of 400 mg administered once daily on a 5 days on, 2 days off weekly schedule. This intermittent regimen achieved a remarkable outcome: it more than doubled the steady-state drug exposure compared to continuous dosing, while simultaneously improving the drug's safety and tolerability profile. This finding was critical, as it demonstrated that higher, more efficacious drug levels could be achieved without a corresponding increase in treatment-related discontinuations, thereby widening the therapeutic index.[16]

4.3. The Cyclin E1 Overexpression Hypothesis: A Predictive Biomarker Strategy

A cornerstone of the Azenosertib clinical development program is the identification and validation of Cyclin E1 as a predictive biomarker of response. The CCNE1 gene, which encodes the Cyclin E1 protein, is a known oncogenic driver in numerous solid tumors, including a significant subset of HGSOC.[1] High levels of Cyclin E1, which can result from gene amplification or other mechanisms, are strongly associated with resistance to conventional chemotherapy and poor patient outcomes.[1]

The biological rationale linking Cyclin E1 to WEE1 inhibitor sensitivity is compelling. Cyclin E1 partners with CDK2 to drive the cell cycle through the G1/S transition. Overexpression of Cyclin E1 leads to hyperactivation of CDK2, which causes uncontrolled and premature firing of DNA replication origins. This results in a state of chronic, high-level replication stress and an accumulation of DNA damage.[1] To survive this self-inflicted genomic stress, these cancer cells become exceptionally dependent on the WEE1-mediated G2/M checkpoint to pause the cell cycle and attempt repairs.

This hypothesis was confirmed in preclinical studies, which showed a significant association between high Cyclin E1 protein expression and sensitivity to Azenosertib.[4] This translational insight was then carried into the clinic, where retrospective analyses of trial data showed that patients with Cyclin E1-positive tumors derived the greatest benefit from Azenosertib, both as a monotherapy and in combination with chemotherapy.[17] This validation of Cyclin E1 as a predictive biomarker has enabled a focused, biomarker-driven clinical strategy, allowing for the selection of patients most likely to respond and de-risking the path to potential regulatory approval.

Section 5: The Azenosertib Clinical Development Program: A Comprehensive Review

The clinical development of Azenosertib is being pursued through a multifaceted strategy, investigating its potential in three distinct therapeutic settings: as a monotherapy, in combination with traditional chemotherapies, and in combination with other molecularly targeted agents.[1] This broad approach aims to maximize the drug's utility across a wide range of oncologic indications with high unmet need.

5.1. Monotherapy Trials in Gynecologic Cancers and Solid Tumors

The monotherapy program is heavily focused on tumors with intrinsic genomic instability, where the synthetic lethal relationship with WEE1 inhibition is most pronounced.

  • DENALI (NCT05128825): This is a pivotal Phase 2, open-label, multicenter study evaluating Azenosertib monotherapy in patients with platinum-resistant high-grade serous ovarian, fallopian tube, or primary peritoneal cancer.[19] A key feature of this trial is its focus on patients with tumors driven by Cyclin E1 overexpression, serving as the primary clinical platform to validate the biomarker strategy.[1]
  • TETON (NCT04814108): This Phase 2, open-label study is assessing the efficacy and safety of Azenosertib in women with recurrent or persistent Uterine Serous Carcinoma (USC).[21] USC is an aggressive subtype of endometrial cancer known for its high degree of genomic instability, making it a strong candidate for WEE1 inhibition.
  • ZN-c3-001 (NCT04158336): This foundational Phase 1 study has been instrumental in the Azenosertib program. It began as a dose-escalation trial in patients with advanced solid tumors to determine safety and identify the maximum tolerated dose. It later evolved to include dose optimization and expansion cohorts, which were critical for establishing the 400 mg intermittent RP2D and for providing the first signals of clinical activity in specific tumor types like USC and ovarian cancer.[22]

5.2. Combination Therapy Trials: Synergistic Approaches

The rationale for combination therapy is to enhance the cytotoxic effects of DNA-damaging agents by preventing cancer cells from repairing the damage induced.

  • MAMMOTH (NCT05198804): This Phase 1/2 study is exploring Azenosertib in patients with platinum-resistant ovarian cancer that is also resistant to PARP inhibitors (PARPi).[1] The trial includes a monotherapy arm to assess Azenosertib's activity in this heavily pretreated population, and originally included combination arms with the PARPi niraparib, based on the strong preclinical rationale for combining DDR inhibitors.
  • Chemotherapy Combinations: Azenosertib is being evaluated with several standard-of-care chemotherapies. A Phase 1b/2 trial (NCT04516447) is assessing its safety and efficacy with paclitaxel, carboplatin, pegylated liposomal doxorubicin (PLD), and gemcitabine in patients with advanced platinum-resistant ovarian cancer.[1] Additionally, a Phase 1/2 trial (NCT04833582) is investigating Azenosertib in combination with gemcitabine for adult and pediatric patients with relapsed or refractory osteosarcoma, a cancer often characterized by TP53 mutations.[1]
  • Molecularly Targeted Agent Combinations: In collaboration with Pfizer, Zentalis initiated a Phase 1/2 trial (NCT05743036) combining Azenosertib with the BEACON regimen (encorafenib and cetuximab) in patients with BRAF V600E-mutated metastatic colorectal cancer (mCRC).[1]

5.3. Analysis of Discontinued and Terminated Studies

Not all investigational paths have proceeded as planned, reflecting the dynamic nature of clinical development and strategic resource allocation.

  • The Phase 1/2 trial in mCRC (NCT05743036) was terminated. The stated reason was a "change in therapeutic landscape," which suggests that evolving standards of care or a strategic prioritization of other indications led to the decision to halt the study.[28]
  • The combination arm of the MAMMOTH trial (NCT05198804) evaluating Azenosertib with niraparib was discontinued. This decision was made because efficacious exposures of Azenosertib could not be achieved in the combination setting, likely due to overlapping toxicities or a prohibitive drug-drug interaction that limited dosing.[25]
  • A Phase 1 study planned to evaluate Azenosertib with bevacizumab and pembrolizumab in solid tumors (NCT05431582) was withdrawn prior to patient enrollment, indicating a strategic decision not to pursue this particular combination at the time.[32]

The following table provides a consolidated overview of the key clinical trials in the Azenosertib development program.

Table 2: Detailed Overview of Key Azenosertib Clinical Trials

Trial Name / IDNCT NumberPhaseIndication(s)InterventionStatus
DENALINCT051288252Platinum-Resistant Ovarian, Fallopian Tube, or Primary Peritoneal Cancer (Cyclin E1 driven)Azenosertib MonotherapyActive
TETONNCT048141082Recurrent/Persistent Uterine Serous CarcinomaAzenosertib MonotherapyActive
ZN-c3-001NCT041583361Advanced Solid TumorsAzenosertib MonotherapyActive
MAMMOTHNCT051988041/2PARP-Resistant Ovarian CancerAzenosertib Monotherapy & Combination with NiraparibActive (Monotherapy); Discontinued (Combination)
ZN-c3-003NCT048335821/2Relapsed/Refractory OsteosarcomaAzenosertib + GemcitabineActive
NCT04516447NCT045164471b/2Platinum-Resistant Ovarian CancerAzenosertib + Chemotherapy (Paclitaxel, Carboplatin, etc.)Active
ZN-c3-016NCT057430361/2BRAF V600E-mutant Metastatic Colorectal CancerAzenosertib + Encorafenib + CetuximabTerminated
NCT05431582NCT054315821Metastatic Solid Tumors (CCNE1 amplified, TP53 mutant)Azenosertib + Bevacizumab ± PembrolizumabWithdrawn

Section 6: Synthesis of Clinical Efficacy and Safety Data

6.1. Efficacy Analysis in Key Indications: Ovarian and Uterine Cancers

Clinical data from the Azenosertib program have demonstrated meaningful and encouraging anti-tumor activity, particularly in heavily pretreated patient populations with high unmet needs.

Platinum-Resistant Ovarian Cancer (PROC): The most compelling efficacy data to date comes from the monotherapy trials in PROC, especially when enriching for the Cyclin E1 biomarker.

  • In the Phase 2 DENALI trial (Part 1b), single-agent Azenosertib administered at the 400 mg intermittent dose yielded a confirmed Objective Response Rate (ORR) of 34.9% (95% CI, 21.0%-50.9%) among 43 response-evaluable patients with Cyclin E1-positive tumors. In the broader intent-to-treat population (n=48), the ORR was 31.3%. The median Duration of Response (DOR) was approximately 5.5 months and was still maturing at the time of data cutoff.[18]
  • Earlier data from the Phase 1 ZN-c3-001 trial also showed strong signals. In a combined cohort of 19 heavily pretreated patients with PROC and USC treated with an intermittent schedule, the confirmed ORR was 36.8%, with a median Progression-Free Survival (PFS) of 6.5 months.[16]

Combination with Chemotherapy in PROC: Azenosertib has also shown strong synergistic activity with standard chemotherapy.

  • In a Phase 1b trial, the combination of Azenosertib with paclitaxel resulted in a confirmed ORR of 50.0% and a median PFS of 7.4 months. The combination with carboplatin yielded an ORR of 35.7% and a median PFS of 10.4 months. In these cohorts, patients with Cyclin E1-positive tumors appeared to derive the most benefit.[17]

Uterine Serous Carcinoma (USC):

  • In the USC expansion cohort of the Phase 1 ZN-c3-001 trial, among 9 evaluable patients treated with doses of 300 mg or higher, the confirmed partial response rate was 33.3%. The overall disease control rate (including stable disease) was an impressive 88.9%, indicating significant clinical activity in this aggressive cancer type.[35]

6.2. Comprehensive Safety and Tolerability Profile Across Studies

Azenosertib has been administered to over 500 patients in monotherapy trials and has been described as generally well-tolerated, with a manageable safety profile, particularly with the optimized intermittent dosing schedule.[22] The most frequently reported treatment-related adverse events (TRAEs) are consistent with the mechanism of action and are primarily hematologic and gastrointestinal in nature.[22]

  • Common TRAEs: Across multiple studies, the most common any-grade TRAEs include nausea, neutropenia, thrombocytopenia, anemia, diarrhea, vomiting, and fatigue.[22]
  • Grade 3 or Higher TRAEs: The most significant toxicities are hematologic. In a multicenter trial combining Azenosertib with chemotherapy (NCT04516447), Grade 3 or higher TRAEs included thrombocytopenia (27.5%), neutropenia (25.5%), and anemia (15.7%).[22] Gastrointestinal events are typically lower grade, with Grade 3 or higher nausea, vomiting, and diarrhea occurring in less than 4% of patients in the same trial.[22]
  • Serious Adverse Events and Clinical Hold: The safety profile is not without significant risks. The program's most serious safety challenge arose in the DENALI trial, where two treatment-related Grade 5 adverse events (deaths) occurred due to presumed sepsis.[18] These events, likely stemming from severe myelosuppression, prompted the FDA to place a partial clinical hold on the monotherapy studies in June 2024. This underscores that while the toxicity profile is considered manageable, it requires careful patient monitoring and proactive management of hematologic side effects to mitigate the risk of severe infections.

The following table summarizes key efficacy and safety outcomes from the most informative clinical trials.

Table 3: Summary of Efficacy and Safety Outcomes from Pivotal Trials

Trial (Cohort)IndicationNEfficacy Outcome(s)Key Grade ≥3 TRAEs (%)Source(s)
DENALI (Part 1b)Cyclin E1+ PROC43 (evaluable)ORR: 34.9%; mDOR: ~5.5 mosFatigue (15.7%), Thrombocytopenia (11.8%), Neutropenia (10.8%)18
ZN-c3-001PROC & USC (intermittent dose)19ORR: 36.8%; mPFS: 6.5 mosNot specified in detail16
ZN-c3-001 (USC Cohort)Uterine Serous Carcinoma9 (evaluable)ORR: 33.3%; DCR: 88.9%Not specified in detail35
NCT04516447Platinum-Resistant Ovarian CancerNot specifiedORR (w/ Paclitaxel): 50.0%; ORR (w/ Carboplatin): 35.7%Neutropenia (25.5%), Thrombocytopenia (27.5%), Anemia (15.7%)17

Section 7: Regulatory and Competitive Landscape

7.1. Regulatory Pathway: Fast Track Designation and Clinical Hold History

The clinical development of Azenosertib has been marked by significant regulatory interactions with the U.S. FDA, reflecting both the promise and the challenges of this novel agent.

In January 2025, the FDA granted Fast Track Designation to Azenosertib for the treatment of patients with Cyclin E1-positive, platinum-resistant epithelial ovarian, fallopian tube, or primary peritoneal cancer.[41] This designation is a critical milestone, as it is intended to facilitate the development and expedite the review of drugs that treat serious conditions and fill an unmet medical need. It underscores the agency's recognition of the high unmet need in this patient population and the potential of Azenosertib, guided by its biomarker strategy, to provide a meaningful clinical benefit.[41]

However, the program also faced a significant challenge in June 2024, when the FDA placed a partial clinical hold on three monotherapy studies: ZN-c3-001, DENALI (NCT05128825), and TETON (NCT04814108).[22] The hold was initiated following reports of two patient deaths in the DENALI trial, which were attributed to presumed sepsis.[40] This action halted new patient enrollment in these trials while the company worked with the agency to conduct a comprehensive safety review.

In a strong positive signal for the program, the FDA lifted the partial clinical hold in September 2024.[44] Zentalis announced that after reviewing the company's complete response, the agency cleared the resumption of enrollment in all ongoing studies with no required changes to the clinical development plan.[45] The successful and relatively swift resolution of the hold without mandated protocol modifications suggests that the FDA was satisfied with the existing safety data and risk mitigation strategies. This outcome, while originating from a negative event, may ultimately reinforce confidence in the manageability of Azenosertib's safety profile when administered to the appropriate patient population with careful monitoring.

7.2. Competitive Analysis of the WEE1 Inhibitor Class

Azenosertib is advancing in a competitive but still nascent field. Crucially, there are no FDA-approved WEE1 inhibitors on the market, giving Azenosertib a clear opportunity to be the first-in-class agent to gain approval.[1]

The most notable historical competitor in the WEE1 inhibitor class is adavosertib (AZD1775). While adavosertib has demonstrated clinical activity in various settings, its development has been challenged by a significant toxicity burden, particularly when combined with chemotherapy, which has hampered its progress.[47] Zentalis has explicitly designed Azenosertib to have advantages over other investigational therapies, citing superior selectivity and pharmacokinetic properties.[1] Indeed, clinical data from Zentalis suggests that Azenosertib combinations have achieved higher response rates and longer progression-free survival compared to historical data for adavosertib combinations in similar patient populations.[17] The superior selectivity of Azenosertib, particularly its weak activity against PLK1, is a key molecular feature intended to translate into a better safety profile and a wider therapeutic window, which could be a decisive competitive advantage.[6]

7.3. Future Clinical and Regulatory Milestones

Zentalis Pharmaceuticals has outlined a clear path forward for the Azenosertib program, with several key milestones anticipated in the near to medium term. The company's strategy is heavily focused on advancing the agent toward a registration-enabling data readout.

  • The initiation of Part 2 of the DENALI trial is a major upcoming step, planned for the first half of 2025.[31] This part of the study is designed with registration intent.
  • Topline data from the complete DENALI Part 2 trial are expected by the end of 2026. If positive, these results have the potential to support an application for accelerated approval from the FDA.[48]
  • Final results from several other key trials, including the Phase 1b monotherapy trial (ZN-c3-001) and the MAMMOTH trial, are anticipated in the second half of 2024.[5]
  • The company has indicated it is on track to submit its first New Drug Application (NDA) for Azenosertib in a gynecologic malignancy in 2026.[51]

Section 8: Expert Analysis and Strategic Outlook

8.1. Strengths and Opportunities in the Azenosertib Program

The Azenosertib development program is built on several key strengths that position it for potential success. The foremost of these is its clear, biologically-driven biomarker strategy centered on Cyclin E1 overexpression. By focusing on a defined patient population with a high unmet need (platinum-resistant ovarian cancer) and a strong biological rationale for response, the program significantly increases its probability of demonstrating a meaningful clinical benefit. This precision-medicine approach is highly favored in modern oncology development and by regulatory agencies.

A second major strength is the successful optimization of the dosing schedule. The transition to an intermittent regimen that enhances both exposure and tolerability represents a critical clinical pharmacology achievement. It appears to have unlocked a more favorable therapeutic index, enabling the administration of a more effective dose while managing toxicity—a common hurdle that has limited other drugs in this class. Finally, as a leading candidate with no approved competitors, Azenosertib has a clear first-in-class opportunity, supported by an expedited regulatory path via its Fast Track Designation. The drug's broad potential for synergistic combinations with a variety of anti-cancer agents, including chemotherapy and potentially antibody-drug conjugates, presents a significant long-term opportunity to expand its use into other indications and lines of therapy.[48]

8.2. Potential Risks, Challenges, and Mitigation Strategies

Despite its promise, the Azenosertib program faces significant risks and challenges. The primary risk remains its safety and tolerability profile. The mechanism of action, which involves disrupting a fundamental cellular process, inherently carries the risk of on-target toxicity, particularly myelosuppression (neutropenia, thrombocytopenia). The two deaths from sepsis that led to the clinical hold are a stark reminder of this risk. While the hold was lifted, successful clinical adoption will depend on the ability of clinicians to carefully monitor patients and proactively manage these hematologic toxicities to prevent severe infections. The therapeutic window, while improved with intermittent dosing, may still be narrow.

Another challenge lies in the execution of the biomarker strategy. The commercial success of Azenosertib in its lead indication will depend on the co-development and regulatory approval of a reliable and accessible companion diagnostic test for Cyclin E1 protein expression. The logistics of implementing this testing in routine clinical practice could present a barrier to uptake. Finally, as with any new targeted therapy, demonstrating a clear and substantial benefit over existing standards of care, which are often less expensive generic chemotherapies, will be essential for securing favorable reimbursement and achieving widespread clinical use.

8.3. Concluding Remarks and Future Trajectory

Azenosertib stands as one of the most promising investigational agents in the field of DNA Damage Response. It is built upon a robust scientific foundation, targeting a validated vulnerability in genomically unstable cancers. The development program, managed by Zentalis Pharmaceuticals, has been characterized by strategic agility, particularly in its pivot to a biomarker-driven approach with Cyclin E1 and its successful optimization of the dosing regimen to enhance the drug's therapeutic index.

The future trajectory of Azenosertib hinges on the successful execution of its late-stage clinical trials. The upcoming registration-intent DENALI Part 2 study will be the ultimate test of its efficacy and safety in the target population of Cyclin E1-positive ovarian cancer. Continued vigilant management of its significant but seemingly predictable safety profile will be paramount.

In conclusion, Azenosertib is a leading candidate to become the first WEE1 inhibitor to reach the market. If the promising efficacy observed in early trials is confirmed in its registrational study, and its safety profile remains manageable in broader use, Azenosertib has the potential to become a transformative new standard of care for a well-defined population of patients with aggressive, difficult-to-treat gynecologic malignancies. Its development serves as a compelling example of modern, biomarker-driven drug development aimed at delivering a highly targeted therapy to the patients most likely to benefit.

Works cited

  1. Pipeline - Zentalis Pharmaceuticals, accessed September 19, 2025, https://www.zentalis.com/pipeline/pipeline
  2. azenosertib | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 19, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=11731
  3. Azenosertib Is a Potent and Selective WEE1 Kinase Inhibitor with Broad Antitumor Activity Across a Range of Solid Tumors - PubMed, accessed September 19, 2025, https://pubmed.ncbi.nlm.nih.gov/40231599
  4. Zentalis Announces Preclinical Data Supporting Cyclin E1 As A Predictive Marker For Azenosertib Treatment At AACR Annual Meeting 2023, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-announces-preclinical-data-supporting-cyclin-e1/
  5. Zentalis Pharmaceuticals Reports Full Year 2023 Financial Results and Operational Updates, accessed September 19, 2025, https://ir.zentalis.com/node/9746/pdf
  6. Azenosertib Is a Potent and Selective WEE1 Kinase Inhibitor with Broad Antitumor Activity Across a Range of Solid Tumors - PubMed Central, accessed September 19, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12314526/
  7. Definition of azenosertib - NCI Drug Dictionary - NCI, accessed September 19, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/azenosertib
  8. Azenosertib | CAS#2376146-48-2 | Wee1 inhibitor - MedKoo Biosciences, accessed September 19, 2025, https://www.medkoo.com/products/48488
  9. ZN-c3 | 2376146-48-2 - ChemicalBook, accessed September 19, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB79751089.htm
  10. Azenosertib (Zn-C3) | Wee1 inhibitor | 99.81%(HPLC) - Selleck ..., accessed September 19, 2025, https://www.selleckchem.com/products/zn-c3.html
  11. Azenosertib (ZN-c3) | Wee1 Inhibitor - MedchemExpress.com, accessed September 19, 2025, https://www.medchemexpress.com/zn-c3.html
  12. Azenosertib - Drug Targets, Indications, Patents - Patsnap Synapse, accessed September 19, 2025, https://synapse.patsnap.com/drug/b6515a3ec85c4b1480ff3f7c6b7edaed
  13. pubmed.ncbi.nlm.nih.gov, accessed September 19, 2025, https://pubmed.ncbi.nlm.nih.gov/40231599#:~:text=Azenosertib%20(ZN%2Dc3)%20is,increases%20in%20DNA%20damage%20markers.
  14. Azenosertib Is a Potent and Selective WEE1 Kinase Inhibitor with Broad Antitumor Activity Across a Range of Solid Tumors - AACR Journals, accessed September 19, 2025, https://aacrjournals.org/mct/article/24/8/1171/763766/Azenosertib-Is-a-Potent-and-Selective-WEE1-Kinase
  15. Azenosertib Is a Potent and Selective WEE1 Kinase Inhibitor with Broad Antitumor Activity Across a Range of Solid Tumors - AACR Journals, accessed September 19, 2025, https://aacrjournals.org/mct/article-pdf/doi/10.1158/1535-7163.MCT-24-1194/3602315/mct-24-1194.pdf
  16. Zentalis Announces Intermittent Azenosertib Monotherapy Dosing Nearly Doubles Efficacy Over Continuous Dosing, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-announces-intermittent-azenosertib-monotherapy-dosing/
  17. Zentalis Announces Presentation of Positive Phase 1b Data Demonstrating Durable Responses and Favorable Safety Profile of Azenosertib in Combination with Chemotherapy at the 2023 ASCO Annual Meeting, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-announces-presentation-positive-phase-1b-data/
  18. Azenosertib Shows Promising Efficacy in Platinum-Resistant Ovarian Cancer, accessed September 19, 2025, https://www.targetedonc.com/view/azenosertib-shows-promising-efficacy-in-platinum-resistant-ovarian-cancer
  19. Azenosertib (ZN-c3) in Subjects With Platinum-Resistant High-Grade Serous Ovarian, Fallopian Tube or Primary Peritoneal Cancer - UCSD Clinical Trials, accessed September 19, 2025, https://clinicaltrials.ucsd.edu/trial/NCT05128825
  20. NCT05128825 | A Study of Azenosertib (ZN-c3) in Subjects With Platinum-Resistant High-Grade Serous Ovarian, Fallopian Tube or Primary Peritoneal Cancer | ClinicalTrials.gov, accessed September 19, 2025, https://clinicaltrials.gov/study/NCT05128825
  21. Clinical Trials - Zentalis Pharmaceuticals, accessed September 19, 2025, https://www.zentalis.com/pipeline/clinical-trials
  22. FDA Places Partial Holds on 3 Trials for Solid Tumor Treatment, Azenosertib, accessed September 19, 2025, https://www.cancernetwork.com/view/fda-places-partial-holds-on-3-trials-for-solid-tumor-treatment-azenosertib
  23. Study Details | NCT04814108 | A Study of ZN-c3 in Women With ..., accessed September 19, 2025, https://clinicaltrials.gov/study/NCT04814108
  24. ZN-c3 in Participants With Solid Tumors – Bladder Cancer Advocacy ..., accessed September 19, 2025, https://bcan.org/clinicaltrials/zn-c3-in-participants-with-solid-tumors/
  25. A Study of ZN-c3 and Niraparib in Subjects With Platinum-Resistant Ovarian Cancer, accessed September 19, 2025, https://clin.larvol.com/trial-detail/NCT05198804
  26. Study Details | NCT04833582 | A Study of ZN-c3 in Combination ..., accessed September 19, 2025, https://clinicaltrials.gov/study/NCT04833582
  27. Zentalis to Present Phase 1 Results of Azenosertib and Gemcitabine in Osteosarcoma at ASCO 2024 - Patsnap Synapse, accessed September 19, 2025, https://synapse.patsnap.com/article/zentalis-to-present-phase-1-results-of-azenosertib-and-gemcitabine-in-osteosarcoma-at-asco-2024
  28. Azenosertib Terminated Phase 1 Trials for Metastatic Colorectal Cancer (CRC) Treatment, accessed September 19, 2025, https://go.drugbank.com/drugs/DB18028/clinical_trials?conditions=DBCOND0134580&phase=1&purpose=treatment&status=terminated
  29. Metastatic Colorectal Cancer (MCRC) Terminated Phase 1 Trials for Azenosertib (DB18028), accessed September 19, 2025, https://go.drugbank.com/indications/DBCOND0039029/clinical_trials/DB18028?phase=1&status=terminated
  30. Study Details | NCT05743036 | ZN-c3 in Adult Participants With ..., accessed September 19, 2025, https://clinicaltrials.gov/study/NCT05743036
  31. Zentalis Pharmaceuticals Shares Updated Clinical Data Demonstrating Meaningful Azenosertib Activity in Cyclin E1+, Platinum-Resistant Ovarian Cancer, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-pharmaceuticals-shares-updated-clinical-data/
  32. Cancer, Ovarian Withdrawn Phase 1 Trials for Azenosertib (DB18028) | DrugBank Online, accessed September 19, 2025, https://go.drugbank.com/indications/DBCOND0063542/clinical_trials/DB18028?phase=1&status=withdrawn
  33. Malignant Tumor of Breast Withdrawn Phase 1 Trials for Azenosertib, accessed September 19, 2025, https://go.drugbank.com/indications/DBCOND0072656/clinical_trials/DB18028?phase=1&status=withdrawn
  34. Azenosertib Monotherapy Appears Safe and Effective in Ovarian Cancer | OncLive, accessed September 19, 2025, https://www.onclive.com/view/azenosertib-monotherapy-appears-safe-and-effective-in-ovarian-cancer
  35. Abstract CT029: Safety and clinical activity of single-agent ZN-c3, an oral WEE1 inhibitor, in a phase 1 trial in subjects with recurrent or advanced uterine serous carcinoma (USC) - ResearchGate, accessed September 19, 2025, https://www.researchgate.net/publication/361320690_Abstract_CT029_Safety_and_clinical_activity_of_single-agent_ZN-c3_an_oral_WEE1_inhibitor_in_a_phase_1_trial_in_subjects_with_recurrent_or_advanced_uterine_serous_carcinoma_USC
  36. Abstract CT029: Safety and clinical activity of single-agent ZN-c3, an oral WEE1 inhibitor, in a phase 1 trial in subjects with recurrent or advanced uterine serous carcinoma (USC) | Cancer Research - AACR Journals, accessed September 19, 2025, https://aacrjournals.org/cancerres/article/82/12_Supplement/CT029/701937/Abstract-CT029-Safety-and-clinical-activity-of
  37. Zentalis Pharmaceuticals Provides Update on Azenosertib Clinical Development Program, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-pharmaceuticals-provides-update-azenosertib-clinical/
  38. Zentalis Pharmaceuticals Announces Positive Initial Clinical Data on ZN-c3, its Wee1 Inhibitor, in Patients with Advanced Ovarian Cancer at AACR, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-pharmaceuticals-announces-positive-initial-clinical/
  39. Azenosertib Monotherapy Yields Encouraging Responses in Cyclin E1+ PROC, accessed September 19, 2025, https://www.cancernetwork.com/view/azenosertib-monotherapy-yields-encouraging-responses-in-cyclin-e1-proc
  40. FDA Places Partial Clinical Holds on 3 Trials Evaluating Azenosertib in Advanced Solid Tumors | OncLive, accessed September 19, 2025, https://www.onclive.com/view/fda-places-partial-clinical-holds-on-3-trials-evaluating-azenosertib-in-advanced-solid-tumors
  41. Zentalis' Azenosertib Gets FDA Fast Track Status For Cyclin E1 Positive Cancer Patients, accessed September 19, 2025, https://www.nasdaq.com/articles/zentalis-azenosertib-gets-fda-fast-track-status-cyclin-e1-positive-cancer-patients
  42. Zentalis Pharmaceuticals Announces Azenosertib Fast Track Designation and Virtual Corporate Event to Present Updated Data from Azenosertib Clinical Studies, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-pharmaceuticals-announces-azenosertib-fast-track/
  43. Zentalis Pharmaceuticals Announces FDA Fast Track Designation for Azenosertib in Cyclin E1 Positive Ovarian Cancer Patients | Nasdaq, accessed September 19, 2025, https://www.nasdaq.com/articles/zentalis-pharmaceuticals-announces-fda-fast-track-designation-azenosertib-cyclin-e1
  44. FDA lifts partial clinical hold on Zentalis Pharma's cancer WEE1 inhibitor, accessed September 19, 2025, https://www.clinicaltrialsarena.com/news/fda-lifts-partial-clinical-hold-on-zentalis-pharmas-cancer-wee1-inhibitor/
  45. Zentalis Pharmaceuticals Announces FDA Has Lifted Partial Clinical Hold on Azenosertib Studies, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-pharmaceuticals-announces-fda-has-lifted-partial/
  46. FDA Lifts Partial Clinical Hold on Trials Evaluating Azenosertib in Advanced Solid Tumors, accessed September 19, 2025, https://www.onclive.com/view/fda-lifts-partial-clinical-hold-on-trials-evaluating-azenosertib-in-advanced-solid-tumors
  47. (PDF) WEE1 Inhibitor: Clinical Development - ResearchGate, accessed September 19, 2025, https://www.researchgate.net/publication/353302205_WEE1_Inhibitor_Clinical_Development
  48. Zentalis Pharmaceuticals Presents Updated Clinical Data at the Society of Gynecologic Oncology 2025 Annual Meeting on Women's Cancer, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-pharmaceuticals-presents-updated-clinical-data-society/
  49. Zentalis Pharmaceuticals Announces First Patient Dosed in DENALI Part 2 Clinical Trial of Azenosertib in Patients with Cyclin E1+ PROC, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-pharmaceuticals-announces-first-patient-dosed-denali/
  50. Zentalis Pharmaceuticals Reports First Quarter 2025 Financial Results and Operational Progress, accessed September 19, 2025, https://ir.zentalis.com/news-releases/news-release-details/zentalis-pharmaceuticals-reports-first-quarter-2025-financial/
  51. A Study of ZN-c3 in Participants With Solid Tumors - larvol clin, accessed September 19, 2025, https://clin.larvol.com/trial-detail/NCT04158336

Published at: September 19, 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.