Small Molecule
C24H25N5O3S
1232416-25-9
Berzosertib is a first-in-class, potent, and selective intravenous inhibitor of the Ataxia telangiectasia and Rad3-related (ATR) kinase. As a pioneer in its therapeutic class, it has been instrumental in validating the strategy of targeting the DNA Damage Response (DDR) in oncology. Its clinical development has yielded a dichotomous narrative: demonstrating significant, practice-informing efficacy in specific, biomarker-enriched populations such as platinum-resistant ovarian cancer, while simultaneously failing to meet endpoints in broader, less-selected patient groups, exemplified by the discontinuation of a pivotal trial in small cell lung cancer (SCLC).
The primary mechanism of action for Berzosertib involves the blockade of the ATR-Checkpoint Kinase 1 (Chk1) signaling pathway, a critical cell cycle checkpoint activated by replication stress. This inhibition prevents cancer cells from repairing damaged DNA, leading to mitotic catastrophe and apoptosis. This therapeutic strategy is based on the principle of synthetic lethality, which is particularly effective in tumors with pre-existing DDR defects, such as those with Ataxia-telangiectasia mutated (ATM) or TP53 mutations.
The most compelling clinical evidence for Berzosertib's efficacy comes from the randomized Phase II trial NCT02595892 in platinum-resistant ovarian cancer. In this study, the addition of Berzosertib to gemcitabine significantly improved progression-free survival compared to gemcitabine alone. Conversely, the global Phase II DDRiver SCLC 250 trial in platinum-resistant SCLC was discontinued for futility, a major setback that underscores the critical importance of patient selection for this class of agents.
The primary and dose-limiting toxicity of Berzosertib, particularly when used in combination with cytotoxic chemotherapy, is myelosuppression, manifesting as anemia, thrombocytopenia, and neutropenia. This hematologic toxicity is a direct on-target effect of ATR inhibition in rapidly dividing hematopoietic progenitor cells and represents the main challenge in achieving a wide therapeutic index.
The future of Berzosertib is contingent on a biomarker-driven development strategy. Its path forward lies not in broad applications but in precision oncology, targeting tumors with validated signatures of DDR deficiency or high replication stress. Ongoing trials with novel combination partners, such as the DNA-damaging agent lurbinectedin and the antibody-drug conjugate sacituzumab govitecan, will be crucial in determining its ultimate place in the therapeutic armamentarium. Its legacy as a foundational ATR inhibitor continues to inform the development of next-generation oral agents in this promising class.
Berzosertib represents a pioneering effort in the clinical application of DNA Damage Response (DDR) inhibition, specifically targeting the ATR kinase. Its journey from an early-stage asset to a widely studied investigational agent reflects the growing interest in exploiting the inherent genomic instability of cancer cells as a therapeutic vulnerability.
Berzosertib was originally invented and developed by Vertex Pharmaceuticals under the development codes VE-822 and VX-970.[1] It emerged from research programs aimed at identifying potent and selective inhibitors of the phosphoinositide 3-kinase-related kinase (PIKK) family. In 2017, the development and commercial rights for the compound were licensed to Merck KGaA, Darmstadt, Germany, which operates as EMD Serono in the United States and Canada.[1] Under Merck KGaA, the drug was assigned the development code M6620 and became the lead candidate in the company's DDR inhibitor portfolio.[3] This transition from a biotechnology innovator to a major global pharmaceutical company signified a strong belief in the therapeutic potential of the ATR inhibitor class and the broader DDR field. Berzosertib holds the distinction of being the first ATR inhibitor to be evaluated in a randomized clinical trial, establishing a clinical precedent for this mechanism of action.[3]
Berzosertib is a synthetic organic small molecule designed for therapeutic use.[7] Its fundamental properties are well-characterized and serve as the basis for its pharmacological activity. The molecule is classified chemically as a sulfonamide and belongs to the class of organic compounds known as benzenesulfonyl compounds, which are defined by an aromatic structure containing a benzene ring that carries a sulfonyl group.[7] Key identifiers and physicochemical properties are summarized in Table 1.
Identifier/Property | Value | Source Snippets |
---|---|---|
Generic Name | Berzosertib | 7 |
DrugBank ID | DB11794 | 1 |
CAS Number | 1232416-25-9 | 1 |
Development Codes | VX-970, VE-822, M6620 | 1 |
Drug Type | Small Molecule | 7 |
IUPAC Name | 3-[4-(methylaminomethyl)phenyl]-1,2-oxazol-5-yl]-5-(4-propan-2-ylsulfonylphenyl)pyrazin-2-amine | 1 |
Chemical Formula | C24H25N5O3S | 1 |
Average Molecular Weight | 463.56 g·mol⁻¹ | 1 |
Canonical SMILES | CNCc1ccc(cc1)c1noc(c1)c1nc(cnc1N)c1ccc(cc1)S(=O)(=O)C(C)C | 8 |
InChIKey | JZCWLJDSIRUGIN-UHFFFAOYSA-N | 1 |
Lipinski's Rule of Five | Yes (0 rules broken) | 7 |
Veber's Rule | No | 7 |
As an investigational agent, Berzosertib has not been approved for any clinical use by regulatory authorities in any jurisdiction.[3] It is currently in Phase II clinical development for the treatment of various solid tumors.[11]
The drug's regulatory history includes a notable designation from the European Medicines Agency (EMA). Berzosertib was granted a rare disease (orphan) designation for the "Treatment of small cell lung cancer".[10] This status is conferred to encourage the development of medicines for rare, life-threatening, or chronically debilitating conditions where a significant benefit over existing therapies is plausible.[12] However, this designation was officially withdrawn on July 18, 2022.[10] This regulatory event was not an isolated administrative decision but a direct consequence of the clinical trial data emerging at the time. The withdrawal followed shortly after the discontinuation of the pivotal Phase II DDRiver SCLC 250 trial in June 2022, which was stopped after an interim analysis revealed a low probability of success.[5] The failure of this key trial removed the evidence base supporting the potential for "significant benefit" required to maintain the orphan designation, demonstrating a clear cause-and-effect relationship between clinical outcomes and regulatory standing. No specific orphan drug designation from the U.S. Food and Drug Administration (FDA) has been noted.[17]
The therapeutic hypothesis for Berzosertib is rooted in the fundamental biology of the DNA Damage Response, a complex network of cellular pathways that sense, signal, and repair DNA lesions to maintain genomic integrity. Berzosertib targets a central node in this network, the ATR kinase, to exploit the unique dependencies of cancer cells.
The Ataxia telangiectasia and Rad3-related (ATR) protein is a master regulator and apical kinase within the DDR network.[10] While other key DDR kinases like ATM and DNA-dependent protein kinase (DNA-PK) primarily respond to DNA double-strand breaks (DSBs), ATR is activated by a wider spectrum of DNA damage, most critically by the presence of extensive regions of single-stranded DNA (ssDNA).[21] Such ssDNA structures are a common feature of replication stress, a state where the DNA replication machinery encounters obstacles, leading to the stalling or collapse of replication forks.[22]
Replication stress is a constitutive feature of many cancers. It is often driven by the expression of oncogenes (e.g., MYC, CCNE1) that promote uncontrolled cell proliferation, overwhelming the normal replication process.[22] To survive this constant endogenous stress, cancer cells become highly dependent on the ATR pathway. Upon sensing ssDNA coated by Replication Protein A (RPA), ATR is recruited and activated. It then phosphorylates a vast array of downstream substrates, with Checkpoint Kinase 1 (Chk1) being one of the most critical.[10] The activated ATR-Chk1 signaling cascade initiates a comprehensive cellular response that includes:
This protective function is particularly vital for cancer cells, which frequently harbor defects in other checkpoint pathways, such as the G1 checkpoint controlled by p53.[22]
Berzosertib is a highly potent and selective ATP-competitive inhibitor of ATR kinase activity.[1] Preclinical characterization has demonstrated a high affinity for its target, with a dissociation constant (
Ki) of less than 0.3 nM and a half-maximal inhibitory concentration (IC50) of approximately 19 nM in cellular assays.[20] Its selectivity is a key feature; it is over 1000-fold more selective for ATR than for DNA-PK and maintains significant selectivity over the closely related ATM kinase (
Ki = 34 nM).[20]
By binding to the kinase domain of ATR, Berzosertib prevents the phosphorylation and subsequent activation of Chk1 and other downstream substrates, effectively shutting down the ATR-mediated signaling cascade.[10] The functional consequence of this molecular inhibition is the abrogation of the DNA damage checkpoint. In the presence of DNA damage (either endogenous or therapy-induced), cells treated with Berzosertib are unable to arrest the cell cycle to perform repairs. They are forced to progress into mitosis with damaged DNA, an event that leads to replication fork collapse, the accumulation of catastrophic DSBs, widespread chromosomal abnormalities, and ultimately, apoptotic cell death.[10]
The central therapeutic strategy underpinning Berzosertib is the concept of synthetic lethality. This principle describes a situation where the loss of function in either of two genes individually is viable, but the simultaneous loss of both is lethal.[22] In the context of Berzosertib, the targeted inhibition of ATR is synthetically lethal with pre-existing defects in other DDR pathways commonly found in cancer cells.
Many tumors exhibit a defective G1 cell cycle checkpoint due to mutations in key tumor suppressor genes like TP53 or ATM. This loss of the primary "gatekeeper" checkpoint makes these cancer cells critically dependent on the ATR-governed intra-S and G2/M checkpoints to manage replication stress and repair DNA damage before cell division.[20] By administering Berzosertib, this remaining essential checkpoint is disabled. The cancer cell, now lacking any functional mechanism to pause and repair, accumulates lethal levels of genomic damage and is selectively eliminated. In contrast, normal, healthy cells typically have an intact G1 checkpoint and lower levels of replication stress, making them less sensitive to the effects of ATR inhibition.[20] This differential dependency creates a therapeutic window. This biological rationale is strongly supported by preclinical findings that demonstrated that defects in the ATM-p53 pathway were predictive of tumor cell sensitivity to Berzosertib.[20]
While Berzosertib has shown some single-agent activity in tumor models with high intrinsic replication stress and DDR defects, its primary therapeutic potential lies in its ability to act as a powerful sensitizing agent, augmenting the efficacy of conventional DNA-damaging cancer therapies.[20]
The mechanism of Berzosertib provides a clear and direct explanation for its most significant clinical toxicity. The ATR pathway is not a cancer-specific process but a fundamental biological mechanism essential for the viability of any cell population undergoing rapid replication. The most prominent example of such a population in the human body is the hematopoietic progenitor cells located in the bone marrow. These cells are in a constant state of proliferation to replenish the body's supply of blood cells and, consequently, experience high levels of intrinsic replication stress. Their survival is highly dependent on a functional ATR pathway to maintain genomic stability during this rapid division. When a systemic ATR inhibitor like Berzosertib is administered, it inevitably affects these healthy, rapidly dividing bone marrow cells in the same way it affects cancer cells. This leads directly to the severe myelosuppression—anemia, thrombocytopenia, and neutropenia—that is consistently observed as the dose-limiting toxicity in clinical trials.[35] This hematologic toxicity is therefore not an off-target or unexpected side effect but a direct, on-target consequence of the drug's fundamental mechanism of action. This reality establishes a narrow therapeutic window between achieving an effective antitumor concentration and causing unacceptable damage to essential healthy tissues.
The pharmacokinetic (PK) profile of Berzosertib, which describes how the body absorbs, distributes, metabolizes, and excretes the drug, has been characterized through a series of preclinical and clinical studies. These studies provide a comprehensive understanding of its disposition and have informed its clinical development and dosing schedules.
Berzosertib is formulated for intravenous (IV) administration and is typically delivered as an infusion.[20] A population pharmacokinetic (PopPK) analysis, which integrated data from 240 patients with advanced cancers across two Phase I studies (NCT02157792 and EudraCT 2013-005100-34), provided a robust model of its behavior in humans. The analysis concluded that the pharmacokinetics of Berzosertib are best described by a two-compartment linear model over the evaluated dose range of 18–480 mg/m².[38] For a typical patient, the estimated clearance (CL) was 65 L/h. The terminal elimination half-life (
t1/2) of the parent drug is approximately 17 to 19.6 hours.[38]
The PopPK model revealed a large volume of distribution, with an estimated central volume of distribution (Vc) of 118 L and a peripheral volume of distribution (Vp) of 1030 L. The intercompartmental clearance, describing the rate of transfer between the central and peripheral compartments, was estimated at 295 L/h.[38] This large volume of distribution indicates that Berzosertib distributes extensively from the plasma into peripheral tissues. This is consistent with preclinical studies in mice, which showed extensive distribution into key tissues such as bone marrow, tumor, thymus, and lymph nodes.[41]
A key aspect of Berzosertib's PK is its nonlinear behavior, which is attributed to the saturation of plasma protein binding. This phenomenon occurs at concentrations that are achieved in clinical trials.[41] At lower doses, a larger fraction of the drug is bound to plasma proteins, limiting its distribution. As the dose increases and binding sites become saturated, the proportion of free, unbound drug in the plasma increases. This leads to less than proportional increases in early plasma concentrations but greater than proportional increases in tissue exposure, as more free drug is available to diffuse into tissues.[41]
Metabolism is the primary mechanism of clearance for Berzosertib. A human mass balance study using radiolabeled [¹⁴C]berzosertib demonstrated that the drug undergoes extensive metabolic transformation.[40] This is supported by the finding that circulating metabolites account for a substantial portion (78%) of the total drug-related material in plasma.[40] Furthermore, the terminal half-life of total radioactivity in plasma (64.3 hours) is significantly longer than that of the unchanged parent drug (19.6 hours), indicating the presence of metabolites that are eliminated more slowly than Berzosertib itself.[40]
In vitro studies have identified the cytochrome P450 enzyme CYP3A4 as the primary mediator of Berzosertib's metabolism.[38] The major circulating metabolite has been identified as
M11. This metabolite is considered pharmacologically inactive and is the most abundant metabolic product found in plasma, accounting for 28.2% of the total drug-related material based on peak concentration and 43.5% based on total exposure (AUC).[40]
The definitive routes of excretion for Berzosertib and its metabolites were established in the human [¹⁴C]berzosertib mass balance study.[40] Over a 14-day collection period, a mean total recovery of 89.5% of the administered radioactive dose was achieved. The data clearly show that elimination occurs predominantly through the hepatobiliary system.
These findings confirm that direct renal excretion of the unchanged parent drug is a minor pathway of elimination (estimated at only 5–6%).[38] The overall disposition of Berzosertib involves extensive CYP3A4-mediated metabolism followed by the excretion of metabolites primarily into the feces.
While early preclinical data suggested that Berzosertib had good potential for penetrating the blood-brain barrier [20], this created a degree of optimism for its use in treating brain metastases and primary brain tumors like glioblastoma (GBM). However, subsequent, more detailed preclinical investigations using patient-derived xenograft models of GBM revealed significant obstacles to achieving therapeutic concentrations in the central nervous system.[43] These studies identified a critical disconnect between the initial promise and the clinical reality of treating CNS tumors.
The primary challenges are twofold. First, Berzosertib is a substrate for active efflux transporters at the BBB, which actively pump the drug out of the brain and back into the bloodstream, severely restricting its net accumulation.[43] Second, the drug exhibits a high degree of binding to brain tissue relative to plasma. This high tissue binding sequesters the drug, leading to very low concentrations of the free, unbound fraction that is pharmacologically active and available to interact with its target.[43] This explains why the potent synergy observed between Berzosertib and the chemotherapy temozolomide in vitro could not be recapitulated in in vivo GBM models. The distribution within intracranial tumors was also found to be heterogeneous; while some drug accumulated in the leaky tumor core, concentrations were sub-therapeutic in the invasive tumor rim and surrounding brain tissue, where the BBB remains intact and where tumor recurrence originates.[43] This is a classic challenge in CNS drug development, where it is not sufficient for a drug to simply cross the BBB; it must achieve and sustain adequate concentrations of its free, active form at the site of action. These pharmacokinetic findings strongly suggest that without novel delivery strategies, the utility of systemically administered Berzosertib for primary brain tumors is likely to be limited, a crucial consideration for future clinical trial design.
The clinical development program for Berzosertib has been extensive, exploring its potential across a wide range of malignancies and in combination with numerous standard-of-care agents. This broad investigation has produced a complex and informative set of results, with notable successes in specific contexts and significant setbacks in others, collectively shaping the current understanding of where ATR inhibition may fit into cancer therapy.
Berzosertib has been evaluated in at least 21 clinical trials, primarily Phase I, I/II, and II studies.[30] The program has investigated Berzosertib both as a monotherapy and, more prominently, as a chemosensitizing or radiosensitizing agent.[7] The most frequently studied indications include malignant solid tumors, small cell lung carcinoma (SCLC), and non-small cell lung carcinoma (NSCLC).[7] A key feature of many of these trials, particularly in later stages, has been the inclusion of biomarker criteria for patient selection, with a focus on tumors harboring alterations in DDR pathway genes such as
ATM and ATR.[30]
The most compelling evidence for the clinical efficacy of Berzosertib has emerged from studies in gynecologic cancers, particularly platinum-resistant high-grade serous ovarian cancer.
The investigation of Berzosertib in SCLC has been a story of initial promise followed by a significant setback.
The efficacy of Berzosertib has been variable across other solid tumor types, further reinforcing the need for patient stratification.
The clinical development of Berzosertib continues, with a strategic focus on novel, rational combinations in biomarker-selected populations. Two active trials are particularly noteworthy:
The clinical journey of Berzosertib provides a powerful illustration of the evolution from a broad, histology-based approach to a more refined, precision medicine strategy. The initial hypothesis—that adding an ATR inhibitor to chemotherapy would be broadly effective in cancers with high replication stress—proved to be overly simplistic. This approach led to clear failures, as seen in the urothelial cancer trial where increased toxicity negated any potential benefit, and in the unselected SCLC population of the pivotal DDRiver trial.[5] In parallel, clear successes emerged in specific, biologically defined contexts: platinum-resistant ovarian cancer, a disease state known for its DDR vulnerabilities, and most compellingly, in tumors with confirmed
ATM mutations.[33] This pattern of results strongly argues that the therapeutic value of Berzosertib is not universal but is unlocked by specific molecular vulnerabilities. The failure in the large SCLC trial may not have been a failure of the drug's mechanism, but rather a failure of the patient selection strategy. Consequently, the future of Berzosertib and the entire ATR inhibitor class depends on moving away from histology-driven trials and toward biomarker-selected "basket" trials that enroll patients based on molecular profiles. The ongoing trials with novel partners in HRD-positive tumors reflect this necessary strategic evolution.[46]
Trial ID | Phase | Cancer Type(s) | Combination Agent(s) | Primary Endpoint(s) | Key Findings / Status | Source Snippets |
---|---|---|---|---|---|---|
NCT02595892 | II | Platinum-Resistant Ovarian Cancer | Gemcitabine | Progression-Free Survival (PFS) | Positive: Met primary endpoint. Median PFS 22.9 vs 14.7 weeks (HR 0.57). OS benefit in biomarker-selected subgroups. | 1 |
DDRiver SCLC 250 (NCT04768296) | II | Platinum-Resistant SCLC | Topotecan | Objective Response Rate (ORR) | Negative: Discontinued due to low probability of success at interim analysis. | 3 |
NCT02487095 | I/II | SCLC & Extrapulmonary Small Cell Cancers | Topotecan | MTD (Ph I), ORR (Ph II) | Positive (PoC): Showed promising ORR of 36% and durable responses, providing rationale for DDRiver 250. | 6 |
JAMA Oncology Trial (Pal et al.) | II | Metastatic Urothelial Carcinoma | Cisplatin + Gemcitabine | PFS | Negative: No improvement in PFS (8.0 vs 8.0 mos). Trend toward inferior OS. Higher toxicity. | 37 |
NCT02595931 | I | Advanced Solid Tumors | Irinotecan | MTD, RP2D | Positive Signal: Manageable safety. Promising activity in ATM-mutant tumors (pancreatic cancer). | 33 |
NCT02157792 | I | Advanced Solid Tumors (incl. TNBC) | Monotherapy, Carboplatin, Cisplatin | Safety, MTD, RP2D | Positive (Early Phase): Well tolerated. Preliminary antitumor activity observed. RP2D established. | 31 |
NCT04802174 | I/II | SCLC, HGNEC | Lurbinectedin | MTD (Ph I), ORR (Ph II) | Ongoing: Actively recruiting. | 45 |
NCT04826341 | I/II | SCLC, HRD+ Cancers | Sacituzumab Govitecan | MTD (Ph I), ORR (Ph II) | Ongoing: Actively recruiting. | 45 |
The safety profile of Berzosertib has been extensively characterized in numerous clinical trials. While generally considered to have a manageable toxicity profile, its use is consistently associated with significant on-target adverse events, particularly when combined with cytotoxic chemotherapy.
Myelosuppression is the most common, clinically significant, and dose-limiting toxicity (DLT) associated with Berzosertib treatment.[20] This is a direct consequence of inhibiting ATR in the rapidly proliferating hematopoietic progenitor cells of the bone marrow. The incidence of Grade 3 or 4 hematologic adverse events is high across nearly all combination regimens studied.
The clinical consequence of this severe myelosuppression is significant. It frequently requires dose reductions, interruptions, or delays in treatment, not only for Berzosertib but also for the backbone chemotherapeutic agent. This was clearly demonstrated in the urothelial cancer trial, where patients receiving the Berzosertib combination received a significantly lower median dose of cisplatin, which may have compromised the regimen's overall efficacy.[35]
Non-hematologic adverse events are also observed, though they are typically less frequent and of lower severity than the hematologic toxicities.
The safety profile of Berzosertib can be viewed as a form of "mechanistic biomarker" that confirms its potent on-target activity. The consistent and severe myelosuppression is a double-edged sword: on one hand, it provides clear evidence of systemic, biologically effective ATR inhibition. On the other hand, it precisely defines the drug's primary therapeutic limitation. The challenge is not that the drug is ineffective, but rather that its mechanism works systemically on all rapidly dividing cells, both cancerous and healthy. This creates a direct, causal link between the desired antitumor effect and the dose-limiting side effect. This reframes the central clinical problem from simply managing side effects to the more complex task of creating a therapeutic window, which might require innovative strategies like intermittent dosing, development of tumor-targeted delivery systems, or co-administration of bone marrow-protective agents.
Data from the European Chemicals Agency (ECHA) C&L Inventory, based on preclinical studies, provide warnings about potential long-term toxicities associated with Berzosertib.[10] These GHS classifications are consistent with a compound that targets a fundamental process of DNA maintenance and cell division:
These preclinical findings underscore the need for careful long-term safety monitoring in patients who receive the drug.
Adverse Event (Grade ≥3) | + Cisplatin / Veliparib 35 | + Irinotecan 33 | + Cisplatin / Gemcitabine 37 | + Topotecan 6 |
---|---|---|---|---|
Anemia | 37.7% | 20% | N/A (not specified) | 42.3% |
Thrombocytopenia | 32.1% | N/A | 59% (vs 39% control) | 57.7% |
Neutropenia / Neutrophil ↓ | 22.6% | 34% | 37% (vs 27% control) | 50.0% |
Leukopenia / WBC ↓ | 24.5% | 28% | N/A | N/A |
Lymphopenia / Lymphocyte ↓ | 20.8% | 30% | N/A | 69.2% |
Diarrhea | N/A | 16% | N/A | N/A |
Fatigue | N/A | 8% | N/A | N/A |
Berzosertib's extensive clinical evaluation has provided invaluable lessons for the entire field of DDR inhibition. Its mixed results have clarified the path forward, emphasizing the need for precision medicine while highlighting the challenges inherent to this therapeutic class. A strategic analysis of its position in the competitive landscape, its core challenges, and its potential future reveals a complex but informative picture.
Berzosertib was the first ATR inhibitor to enter the clinic, but it is no longer the only one. Several other agents are now in development, creating a competitive landscape.[22]
The clinical journey of Berzosertib has been defined by one overarching challenge: achieving a sufficiently wide therapeutic window.
The future of the Berzosertib program depends on its ability to adapt to these lessons. The strategy is now shifting toward smarter, more rational combinations in highly selected patient populations.
Berzosertib is a landmark molecule in the development of DNA Damage Response inhibitors. It has successfully validated ATR as a druggable and clinically relevant target in oncology, demonstrating that exploiting the principle of synthetic lethality can lead to profound and durable responses in some patients with hard-to-treat cancers.
However, its clinical development has also been a cautionary tale, illustrating the quintessential challenges of targeted therapy. Its potent, on-target mechanism is a double-edged sword, leading to dose-limiting myelosuppression that narrows the therapeutic window and complicates combination strategies. The divergent outcomes of its clinical trials—from the clear success in biomarker-selected, platinum-resistant ovarian cancer to the definitive failure in unselected small cell lung cancer—have delivered an unequivocal message: the future of ATR inhibition lies in precision oncology.
The ultimate clinical role of Berzosertib may be as a niche therapeutic for specific, molecularly defined patient populations, such as those with ATM-deficient tumors. Its greater and more enduring legacy, however, will be the foundational clinical and biological knowledge it has generated. The lessons learned from the successes and failures of Berzosertib have illuminated the path forward, paving the way for a new generation of more refined, biomarker-guided, and potentially oral ATR inhibitors that may one day become a standard of care.
Published at: September 15, 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.