AZD-9574, an investigational, orally bioavailable small molecule, is emerging as a significant development in the landscape of oncology therapeutics, specifically within the class of poly (ADP-ribose) polymerase (PARP) inhibitors.[1] Its development is propelled by the persistent need for PARP inhibitors with optimized pharmacological profiles, particularly enhanced selectivity for the PARP1 isoenzyme and the crucial ability to penetrate the blood-brain barrier (BBB) to access the central nervous system (CNS).[3] The rationale underpinning this development is the potential for such characteristics to translate into improved efficacy in challenging malignancies, including primary CNS tumors and brain metastases, alongside a more favorable safety profile compared to earlier-generation, non-selective PARP inhibitors.[3]
The strategic focus on these specific attributes—BBB penetration and PARP1 selectivity—addresses well-documented limitations of existing PARP inhibitors. First-generation agents, while demonstrating clinical utility, are generally hampered by poor CNS distribution and can be associated with hematological toxicities, partly attributed to their broader inhibition of PARP family members, including PARP2.[3] Brain metastases represent a frequent and devastating complication in various cancers, notably in homologous recombination repair (HRR) deficient breast cancer, and primary brain tumors like glioblastoma (GBM) continue to have dismal prognoses.[3] AZD-9574's specific engineering for BBB penetration [1] and its high selectivity for PARP1 aim to directly address these unmet needs.[3] This dual optimization signifies a considerable advancement in the PARP inhibitor class, positioning AZD-9574 not merely as an incremental improvement but as a molecule designed to fill critical therapeutic gaps in the management of difficult-to-treat cancers.
AZD-9574 is currently under investigation for the treatment of advanced solid malignancies. A particular emphasis is placed on tumors characterized by defects in DNA damage response (DDR) pathways, such as those exhibiting HRR deficiencies, including mutations in genes like BRCA1 and BRCA2.[3] AstraZeneca's oncology strategy involves attacking cancer from multiple angles, and AZD-9574 represents a key component of its efforts to advance the field of PARP inhibition.[10]
AZD-9574 is being developed by AstraZeneca.[8] The investigational drug is identified by the code AZD-9574 and has been assigned the International Nonproprietary Name (INN) Palacaparib.[1] Other chemical and regulatory identifiers include CAS Registry Number 2756333-39-6, UNII 9UG32UQW48, ChEMBL ID CHEMBL5095223, and NCI Thesaurus Code C188237.[1]
The development of AZD-9574, alongside other DDR-targeted agents within AstraZeneca's portfolio such as AZD5305 (another PARP1-selective inhibitor) and ceralasertib (an ATR inhibitor), highlights the company's sustained and profound strategic commitment to the DDR pathway as a central pillar of its oncology research and development.[10] This focused effort suggests that AZD-9574 is not an isolated endeavor but rather part of a broader, evolving strategy. This strategy likely aims to refine and expand the therapeutic applications of DDR-targeting drugs, leveraging insights and experiences gained from earlier agents in this class and reflecting a commitment to next-generation therapies with potentially superior efficacy and safety profiles.
AZD-9574 functions as a potent and highly selective inhibitor of Poly(ADP-ribose) Polymerase 1 (PARP1).[1] PARP enzymes, with PARP1 being a key member, are integral to cellular DNA repair mechanisms, particularly the base excision repair (BER) pathway, which addresses DNA single-strand breaks (SSBs).[1]
A distinguishing characteristic of AZD-9574 is its remarkable selectivity for PARP1 over other PARP isoenzymes. Preclinical data indicate a selectivity of greater than 8000-fold for PARP1 compared to PARP2, and also high selectivity over PARP3, PARP5a, and PARP6.[3] This contrasts sharply with first-generation PARP inhibitors, which typically exhibit non-selective inhibition of both PARP1 and PARP2.[3]
The anti-neoplastic activity of AZD-9574 is mediated through a dual mechanism: it not only catalytically inhibits PARP1 enzymatic activity but, critically, it also "traps" PARP1 molecules at the sites of DNA SSBs.[3] These trapped PARP1-DNA complexes are highly cytotoxic. During DNA replication, these complexes can lead to the stalling of replication forks and the conversion of SSBs into more deleterious DNA double-strand breaks (DSBs).
In cancer cells that possess deficiencies in the HRR pathway—for instance, due to loss-of-function mutations in genes such as BRCA1 or BRCA2—these therapy-induced DSBs cannot be repaired efficiently. This accumulation of irreparable DNA damage triggers genomic instability, cell cycle arrest, and ultimately, programmed cell death (apoptosis), a concept known as synthetic lethality.[3] The degree of PARP trapping, beyond simple catalytic inhibition, is increasingly recognized as a pivotal determinant of both the efficacy and, potentially, the toxicity of PARP inhibitors. AZD-9574's characterization as a PARP1-selective trapper suggests an optimization for this more potent mechanism of action. The high selectivity for PARP1 aims to focus this trapping effect on the desired target while mitigating potential toxicities associated with the trapping of other PARP isoforms, particularly PARP2, on DNA. This refined mechanism suggests a sophisticated drug design approach aimed at maximizing a potent anti-cancer effect while minimizing collateral damage, potentially leading to a wider therapeutic window.
The rationale for pursuing PARP1 selectivity is compelling: PARP1 inhibition alone has been demonstrated to be sufficient for inducing synthetic lethality in HRR-deficient cancer cells. Conversely, the inhibition of PARP2 has been implicated in contributing to hematologic toxicities observed with non-selective PARP inhibitors.[3] Therefore, the selective targeting of PARP1 by AZD-9574 aims to maintain or even enhance anti-tumor efficacy while offering an improved safety profile.
Preclinical studies have demonstrated that AZD-9574 effectively inhibits the enzymatic activity of PARP1, known as PARylation. In various cell lines, AZD-9574 exhibited IC50 values for PARylation inhibition typically in the low nanomolar range, between 0.3 and 2 nM, and this effect was observed irrespective of the HRR status of the cells.[7] Comparative studies in A549 cells indicated that AZD-9574 (IC50 of 1.5 nmol/L) was more potent at inhibiting PARylation than olaparib (IC50 of 14.7 nmol/L).[3] Further confirming its PARP1 selectivity, AZD-9574 did not achieve PARylation inhibition in PARP1 knockout (PARP1-/-) cells at concentrations up to 30 µmol/L, whereas it remained effective in PARP2 knockout (PARP2-/-) cells with an IC50 of 1.5 nmol/L.[3]
In vivo target engagement has also been confirmed. In orthotopic glioblastoma (GBM) patient-derived xenograft (PDX) models, administration of AZD-9574 resulted in a greater than 50% reduction in PARylation levels within the tumors, and this target modulation was sustained for up to 24 hours post-dose.[3] Such potent low nanomolar IC50 values for PARylation inhibition and evidence of sustained target engagement in vivo provide a strong preclinical foundation for achieving therapeutic concentrations and effective target modulation in patients, potentially at well-tolerated doses. This robust preclinical pharmacodynamic profile is crucial for successful clinical translation and likely informed the dose selection and escalation strategies in the ongoing CERTIS1 clinical trial.[8] The ability to achieve this level of target engagement with a PARP1-selective and BBB-penetrant agent is a cornerstone of its therapeutic potential. Pharmacodynamic assessments are integral components of the CERTIS1 trial (NCT05417594) to confirm these effects in human subjects.[8]
AZD-9574 is formulated for oral administration and exhibits good bioavailability.[1] A defining and highly significant pharmacokinetic property of AZD-9574 is its engineered ability to efficiently penetrate the BBB and achieve substantial concentrations within the CNS.[1] This characteristic has been extensively validated in preclinical models.
In vivo studies conducted in mice, rats, and cynomolgus monkeys have consistently demonstrated effective BBB penetration.[3] The unbound drug partition coefficient between brain and plasma (Kpuu), a key metric for CNS exposure, was determined to be approximately 0.31 in rats, 0.17 in mice, and notably high at 0.79 in cynomolgus monkeys.[3] These values are significantly greater than those observed for first-generation PARP inhibitors, such as olaparib, which has a Kpuu of less than 0.02.[3]
Positron Emission Tomography (PET) studies using 11C-radiolabeled AZD-9574 in cynomolgus monkeys provided direct visual evidence of clear and significant distribution of the drug into the brain. In stark contrast, PET imaging with 11C-labeled olaparib and 11C-labeled niraparib showed minimal to negligible signal in the brain region, underscoring the superior CNS penetration of AZD-9574.[3] Furthermore, mass spectrometry imaging (MSI) techniques have confirmed homogenous distribution of AZD-9574 within both naive and tumor-bearing brains, with evidence of preferential accumulation within the tumor compartment compared to adjacent normal brain tissue.[3]
The enhanced CNS penetration of AZD-9574 is attributed, in part, to its design for low susceptibility to efflux by active transporter proteins, such as P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP), which are highly expressed at the BBB and often limit the brain distribution of many xenobiotics.[3] The effective BBB penetration by a potent PARP1-selective inhibitor like AZD-9574 has the potential to fundamentally alter the treatment landscape for CNS tumors, such as GBM, and brain metastases. These are indications where systemic therapies, including older PARP inhibitors, have largely been ineffective due to inadequate drug delivery across the BBB.[3] AZD-9574 therefore represents a significant opportunity to apply the established efficacy of PARP inhibition directly within the CNS, addressing a critical unmet medical need.
Comprehensive pharmacokinetic assessments in humans, including detailed characterization of its absorption, distribution, metabolism, and excretion, are primary objectives of the ongoing CERTIS1 clinical trial (NCT05417594).[8]
AZD-9574 (Palacaparib) is a small molecule with defined chemical characteristics. Its molecular formula is C21H22F2N6O2, corresponding to a molecular weight of 428.4 g/mol.[1] The International Union of Pure and Applied Chemistry (IUPAC) name for AZD-9574 is 6-fluoro-5-[(5-fluoro-2-methyl-3-oxo-4H-quinoxalin-6-yl)methyl]piperazin-1-yl]-N-methylpyridine-2-carboxamide.[1] Its structure is further defined by the SMILES notation: CC1=NC2=C(C(=C(C=C2)CN3CCN(CC3)C4=C(N=C(C=C4)C(=O)NC)F)F)NC1=O.[1] Preclinical evaluations have described its overall physicochemical properties as acceptable for drug development in various animal species.[11]
Table 1 provides a summary of key chemical and pharmacological properties of AZD-9574.
Table 1: Key Chemical and Pharmacological Properties of AZD-9574 (Palacaparib)
Property | Details | Reference(s) |
---|---|---|
Drug Name(s) | AZD-9574, Palacaparib | 1 |
Developer | AstraZeneca | 8 |
CAS Registry Number | 2756333-39-6 | 1 |
Molecular Formula | C21H22F2N6O2 | 1 |
Molecular Weight | 428.4 g/mol | 1 |
IUPAC Name | 6-fluoro-5-[(5-fluoro-2-methyl-3-oxo-4H-quinoxalin-6-yl)methyl]piperazin-1-yl]-N-methylpyridine-2-carboxamide | 1 |
Mechanism of Action | Orally bioavailable, PARP1-selective inhibitor and PARP1-DNA trapper | 1 |
Key Selectivity | PARP1 vs PARP2: >8000-fold | 3 |
BBB Penetration Metrics | Kpuu rat: ~0.31; Kpuu mouse: ~0.17; Kpuu monkey: ~0.79. Qualitatively described as high CNS penetration. | 3 |
The preclinical in vitro evaluation of AZD-9574 has provided robust evidence of its potent and selective activity against PARP1. Its selectivity was rigorously demonstrated across multiple assay platforms, including fluorescence anisotropy, PARylation inhibition assays, and PARP-DNA trapping assays.[3] Biochemical assays confirmed that AZD-9574 exhibits over 8000-fold selectivity for PARP1 compared to PARP2, and also substantial selectivity against other PARP family members such as PARP3, PARP5a, and PARP6.[3] This consistent demonstration of PARP1 selectivity across diverse biochemical and cellular methodologies provides a high degree of confidence in this fundamental characteristic of AZD-9574. Such multi-assay validation strengthens the hypothesis that its pharmacological activity is indeed primarily mediated through PARP1, which in turn supports the potential for an improved therapeutic index by minimizing PARP2-related off-target effects.
In terms of enzymatic inhibition, AZD-9574 potently inhibited PARP1 activity, with IC50 values for PARylation inhibition in the range of 0.3 to 2 nM across various cell lines, an effect observed regardless of their HRR status.[7] Compared to olaparib (IC50 of 14.7 nmol/L), AZD-9574 was more potent in inhibiting PARylation in A549 cells (IC50 of 1.5 nmol/L).[3] The PARP1-selectivity was further underscored in experiments using knockout cell lines: AZD-9574 did not inhibit PARylation in PARP1-/- cells, but effectively inhibited it in PARP2-/- cells, confirming its specificity for PARP1.[3]
Beyond catalytic inhibition, AZD-9574 was shown to trap PARP1 onto DNA in a concentration-dependent manner, with no significant PARP2 trapping observed.[3] This selective trapping is a critical component of its cytotoxic mechanism.
The antiproliferative effects of AZD-9574 were particularly pronounced in cancer cell lines with HRR deficiencies. It selectively inhibited the growth of BRCA-mutant (BRCAm) cell lines.[11] Colony formation assays demonstrated significantly higher potency and selectivity towards HRD-positive (HRD+) models, such as DLD1 BRCA2-/-, SKOV-3 BRCA2-/-, and SKOV-3 PALB2-/- cells.[13] For instance, in DLD1 BRCA2-/- cells, AZD-9574 had an IC50 of 1.38 nM, compared to an IC50 greater than 40 µM in the corresponding BRCA2 wild-type cells, representing an approximately 20,000-fold greater efficacy in the HRD+ context.[13] Similarly potent antiproliferative effects were observed in other BRCA1 mutant cell lines, including MDA-MB-436 (IC50 2 nmol/L) and UWB1.289 (IC50 4.7 nmol/L).[3] Engineered BRCA2-/- cells also showed superior sensitivity to AZD-9574 compared to their BRCA2+/+ counterparts, with a 5 log-fold greater selectivity.[3] The cell lines utilized in these studies were appropriately authenticated and validated to be free of Mycoplasma contamination, ensuring the reliability of the findings.[3]
The promising in vitro activity of AZD-9574 translated into significant antitumor effects in preclinical animal models. As a monotherapy, AZD-9574 demonstrated potent single-agent efficacy in models characterized by HRR deficiency.[3] In a subcutaneous xenograft model using the BRCA1-mutant MDA-MB-436 human breast cancer cell line, daily oral administration of AZD-9574 resulted in dose-dependent tumor regressions. Specifically, tumor regressions of 88% at 30 mg/kg, 86% at 10 mg/kg, and 71% at 3 mg/kg were observed, and these anti-tumor effects were notably durable even after treatment withdrawal.[3]
A critical aspect of AZD-9574's preclinical evaluation was the assessment of its efficacy in CNS tumor models, directly testing the therapeutic relevance of its BBB penetration. In an intracranial xenograft model employing luciferase-tagged MDA-MB-436 cells (a model of breast cancer brain metastases), AZD-9574 administered at doses of 3, 10, and 30 mg/kg once daily led to significant tumor growth suppression and a marked extension in the survival of tumor-bearing mice.[11] Even at a dose of 3 mg/kg, treatment of animals with established intracranial lesions resulted in sustained tumor growth inhibition and a statistically significant increase in survival.[3] This demonstration of substantial anti-tumor activity and survival benefit in orthotopic, intracranial xenograft models serves as a crucial validation of AZD-9574's design as a BBB-penetrant PARP inhibitor. It effectively bridges the gap between demonstrating physical brain presence (pharmacokinetics) and achieving a tangible therapeutic effect (pharmacodynamics and efficacy) within the challenging microenvironment of the CNS. These findings provide a strong preclinical proof-of-concept that AZD-9574 can not only reach CNS tumors but also effectively inhibit their growth, positioning it as a highly promising candidate for primary brain cancers and metastatic disease to the brain.
The therapeutic potential of AZD-9574 has also been explored in combination regimens, most notably with temozolomide (TMZ), a standard-of-care alkylating chemotherapy agent for gliomas. Preclinical studies revealed strong synergistic interactions between AZD-9574 and TMZ in O6-methylguanine-DNA methyltransferase (MGMT)-methylated glioma cell lines, such as SJ-G2 and GBM39 cells. This synergy was observed at nanomolar concentrations of AZD-9574 (30 to 300 nmol/L).[3] The mechanistic rationale for this synergy lies in TMZ's ability to induce DNA lesions, particularly O6-methylguanine (O6-MeG). In MGMT-methylated tumors, where the MGMT repair protein is silenced, these O6-MeG lesions persist and can lead to the formation of SSBs during DNA replication, thereby creating substrates for PARP enzymes and sensitizing cells to PARP inhibitors.[3]
In vivo, the combination of AZD-9574 and TMZ demonstrated enhanced anti-tumor activity. In a subcutaneous SJ-G2 glioma xenograft model, the addition of AZD-9574 to TMZ treatment resulted in significantly greater tumor growth inhibition than TMZ alone.[3] More compellingly, in an orthotopic GBM PDX model (GBM39), the combination of AZD-9574 (at both low, 0.3 mg/kg, and high, 3 mg/kg, doses) with TMZ (12.5 mg/kg) led to a significant increase in the survival of tumor-bearing mice compared to TMZ monotherapy.[3]
A particularly noteworthy finding from these preclinical combination studies was the observation that a higher dose of AZD-9574 (3 mg/kg) combined with a lower dose of TMZ (6.25 mg/kg) provided a survival benefit that was comparable to, or even better than, that achieved with the full standard dose of TMZ (12.5 mg/kg) alone.[3] This suggests that AZD-9574 may enable a reduction in the TMZ dosage without compromising anti-tumor efficacy. If this finding translates to the clinical setting, it could have profound implications for glioma patients by potentially mitigating the often severe dose-dependent toxicities associated with TMZ, such as myelosuppression. This could improve patients' quality of life, enhance treatment compliance, and offer a better-tolerated regimen for a patient population in urgent need of improved therapeutic options. This potential for TMZ dose de-escalation represents a key clinical benefit beyond simply augmenting the anti-tumor effect.
The preclinical safety profile of AZD-9574, particularly concerning hematotoxicity, has been investigated, aligning with the hypothesis that its PARP1 selectivity would confer safety advantages. As a monotherapy, AZD-9574 was reported to be well-tolerated in rats, with no significant adverse effects on hematological parameters. This observation is consistent with the anticipated improved hematotoxicity profile for PARP1-selective inhibitors, as PARP2 inhibition has been strongly implicated as a driver of such toxicities with non-selective agents.[3]
In combination with TMZ, a potentiation of TMZ-induced hematotoxicity was observed, affecting the red cell lineage, neutrophils, and platelets. However, these preclinical studies indicated that such combination-related hematotoxic effects were more effectively mitigated by reducing the dose of TMZ rather than the dose of AZD-9574.[3]
Further supporting a potentially favorable safety margin, in vitro bone marrow assays using human hematopoietic stem and progenitor cells (HSPCs) showed that AZD-9574 had an IC50 of 4.13 µmol/L for reducing HSPC viability. This value indicates that AZD-9574 was less potent in causing hematopoietic toxicity (i.e., potentially safer) compared to the non-selective PARP inhibitor olaparib, which had an IC50 of 0.75 µmol/L in the same assay.[3] Moreover, AZD-9574 demonstrated less potentiation of TMZ-driven hematotoxicity than olaparib at matched concentration ranges in this in vitro system.[3] Some comparisons with another PARP1 selective inhibitor, AZD5305, suggested AZD-9574 had less hematological toxicity in rats, although a third compound, HH102007, reportedly showed even less in specific preclinical settings.[17] It is known that TMZ combinations with some non-selective PARP inhibitors can be overtly toxic.[18]
These direct comparative preclinical data, particularly the lower toxicity to human hematopoietic cells relative to olaparib and the good single-agent tolerability in rats, provide tangible evidence supporting the hypothesis that PARP1 selectivity can indeed translate into a quantifiable improvement in the safety margin. This is a critical factor for successful clinical development, impacting not only patient quality of life but also the feasibility of maintaining adequate dose intensity and enabling more effective and tolerable combination therapies, especially with other myelosuppressive agents.
Table 3 summarizes key preclinical efficacy findings for AZD-9574.
Table 3: Preclinical Efficacy Highlights of AZD-9574
Category | Finding | Reference(s) |
---|---|---|
In Vitro | ||
PARP1 Enzymatic Inhibition IC50 | 0.3-2 nM (various cell lines) | 7 |
PARylation Inhibition IC50 | 1.5 nmol/L (A549 cells) (vs. 14.7 nmol/L for olaparib) | 3 |
Colony Formation IC50 | 1.38 nM (DLD1 BRCA2-/-; >20,000-fold selectivity vs. WT) | 13 |
Colony Formation IC50 | 2 nmol/L (MDA-MB-436, BRCA1m) | 3 |
PARP1 Trapping | Demonstrated PARP1-selective trapping | 3 |
In Vivo (Subcutaneous Xenografts) | ||
MDA-MB-436 (BRCA1m) | Dose-dependent tumor regressions (e.g., 88% at 30 mg/kg QD) | 3 |
In Vivo (Intracranial Xenografts) | ||
MDA-MB-436 luc (BRCA1m) | Dose-dependent tumor regression, significant survival extension (e.g., at 3 mg/kg QD) | 3 |
GBM39 (Orthotopic PDX, with TMZ) | Significant survival extension vs. TMZ alone; TMZ dose reduction possible with AZD-9574 | 3 |
AZD-9574 (Palacaparib) is currently advancing through early-phase clinical development, primarily as a Phase I/IIa investigational agent.[3] The cornerstone of its clinical program is the NCT05417594 study, designated CERTIS1.[9] According to AstraZeneca's pipeline information, AZD-9574 transitioned into Phase II development for advanced solid malignancies around the fourth quarter of 2023.[12] Independent tracking also indicates a Phase II status for gastric cancer.[22]
The overarching hypothesis driving the clinical investigation of AZD-9574 is that its unique pharmacological properties—notably its PARP1 selectivity and ability to penetrate the BBB—will translate into a more efficacious and less toxic therapeutic option compared to currently approved PARP inhibitors. This is particularly anticipated for patients with CNS malignancies (both primary and metastatic) and other HRD+ tumors.[4] The progression to Phase II and the comprehensive, multi-faceted design of the CERTIS1 trial suggest that the preclinical data package for AZD-9574 was robust, prompting an ambitious clinical development strategy by AstraZeneca. This strategy appears aimed at rapidly defining the drug's therapeutic role across a spectrum of tumor types and in various combination regimens, reflecting a high level of confidence in its potential and a desire to quickly identify promising clinical niches.
The CERTIS1 trial (NCT05417594) is formally titled: "A Modular Phase I/IIa, Open-label, Multi-centre Study to Assess the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics and Preliminary Efficacy of Ascending Doses of AZD9574 as Monotherapy and in Combination With Anti-cancer Agents in Patients With Advanced Solid Malignancies".[4] This Phase I/IIa study is sponsored by AstraZeneca.[7] As of the latest available information, the trial is actively recruiting participants, having commenced in June 2022, with an estimated primary completion date of February 2027.[9]
The primary objectives of CERTIS1 are to evaluate the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of AZD-9574, administered both as a monotherapy and in combination with other anti-cancer agents. A key component of the initial phase is dose escalation to determine the optimal dosing regimen(s).[8] Preliminary anti-tumor efficacy will also be assessed.
A distinctive feature of CERTIS1 is its modular design, allowing for the concurrent investigation of AZD-9574 in various contexts [8]:
The trial aims to enroll approximately 490 to 535 participants across its various modules.[8] This complex, modular structure, with its heavy emphasis on biomarker stratification (e.g., HRD status, IDH mutations, HER2 status) and the early exploration of multiple novel combination therapies, reflects a sophisticated precision oncology approach. It suggests an understanding that PARP inhibitors, including next-generation agents like AZD-9574, are likely to offer the greatest benefit in molecularly defined patient populations and potentially through synergistic interactions with other therapies. This trial design aims to maximize the chances of identifying specific niches and regimens where AZD-9574 can provide significant clinical value.
Table 2 provides an overview of the CERTIS1 clinical trial.
Table 2: Overview of the CERTIS1 Clinical Trial (NCT05417594) for AZD-9574
Feature | Details | Reference(s) |
---|---|---|
Trial ID | NCT05417594 | 9 |
Official Title | A Modular Phase I/IIa, Open-label, Multi-centre Study to Assess the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics and Preliminary Efficacy of Ascending Doses of AZD9574 as Monotherapy and in Combination With Anti-cancer Agents in Patients With Advanced Solid Malignancies (CERTIS1) | 9 |
Sponsor | AstraZeneca | 7 |
Phase | I/IIa | 3 |
Current Status | Recruiting | 9 |
Start Date | June 2022 | 9 |
Est. Primary Completion | February 2027 | 9 |
Key Modules/Arms | Mod 1: Monotherapy (Adv. Solid Tumors; specific HRD+ Breast Cancer cohorts +/- Brain Mets) <br> Mod 2: + Temozolomide (IDH-mutant Glioma) <br> Mod 3: PET Sub-study (HRD+ Tumors; IDH-mutant Glioma) <br> Mod 4: + Trastuzumab Deruxtecan (HER2+ Solid Tumors) <br> Mod 5: + Datopotamab Deruxtecan (Various Adv. Solid Tumors incl. TNBC, Endometrial, Ovarian, CRPC) | 8 |
Broad Conditions | Advanced Solid Malignancies | 9 |
Specific Patient Populations (Examples) | BRCA1/2m, PALB2m, RAD51Cm/Dm Ovarian, Breast, Pancreatic, Prostate Cancers; IDH-mutant Glioma; HER2+ Solid Tumors | 8 |
Primary Objectives | Safety, Tolerability, Pharmacokinetics (PK), Pharmacodynamics (PD), Determination of Optimal Dose(s), Preliminary Efficacy | 8 |
The CERTIS1 trial enrolls adult patients (18 years and older) with advanced solid malignancies that have recurred or progressed following prior treatments.[8] Eligible patients generally must have an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 and adequate organ and marrow function.[9]
The specific malignancies and patient populations targeted within the various modules of CERTIS1 are diverse, reflecting the broad therapeutic hypotheses for AZD-9574:
Beyond CERTIS1, development plans also list gastric cancer, non-small cell lung cancer (NSCLC), bladder cancer, colorectal cancer, cervical cancer, endometrial cancer, metastatic biliary tract cancer, vaginal cancer, and vulvar cancer as areas of investigation for AZD-9574.[8]
The patient selection criteria within CERTIS1 strategically integrate two fundamental scientific rationales for AZD-9574. Firstly, the well-established principle of synthetic lethality in HRR-deficient tumors is addressed by targeting cancers with mutations in BRCA1/2, PALB2, and other related genes. Secondly, the unique BBB-penetrant property of AZD-9574 is leveraged by including cohorts specifically for CNS malignancies (IDH-mutant glioma) and tumors with a high propensity for, or existing, brain metastases (e.g., HER2-negative breast cancer with brain metastases). This dual-pronged approach to patient selection is designed to maximize the potential for observing positive clinical signals in populations where one or both of AZD-9574's distinguishing features are most likely to confer a therapeutic benefit.
Within the CERTIS1 trial, AZD-9574 is being evaluated both as a monotherapy and in several combination regimens. AZD-9574 is administered orally in all settings.[8]
The selection of these combination partners is mechanistically driven. PARP inhibitors are known to potentiate the effects of DNA-damaging agents by preventing the repair of lesions induced by these partners.[1] TMZ, which itself penetrates the BBB, is a logical partner for the BBB-penetrant AZD-9574 in glioma, with strong preclinical synergy demonstrated. The ADCs T-DXd and Dato-DXd induce DNA damage via their topoisomerase I inhibitor payloads, creating a rationale for synergy with PARP inhibition. AstraZeneca has a significant strategic focus on ADCs, and preclinical data has already shown promising activity for the combination of Enhertu (T-DXd) with PARP1-selective inhibitors.[10] This combination strategy, therefore, aims to exploit AZD-9574's ability to sensitize tumor cells to DNA damage induced by both established chemotherapy and cutting-edge ADCs, potentially broadening its therapeutic utility.
As is typical for Phase I/IIa oncology trials, the primary objectives of CERTIS1 revolve around establishing the safety and tolerability of AZD-9574.[4] This involves meticulous monitoring and characterization of:
Pharmacokinetic (PK) parameters (to understand drug absorption, distribution, metabolism, and excretion) and pharmacodynamic (PD) markers (to assess target engagement and biological effects) are also key primary or secondary objectives.[8]
Preliminary efficacy will be evaluated using standard oncologic measures, including:
Exploratory biomarker analyses are also planned, with participants consenting to provide tumor tissue and blood samples for correlative studies to identify predictors of response or resistance.[8] While safety and MTD determination are paramount in the initial stages, any early indications of clinical activity, particularly within the CNS-focused cohorts (e.g., glioma, breast cancer with brain metastases), will be of immense interest. Given the high unmet medical need in these areas and AZD-9574's specific design attributes, positive efficacy signals in these challenging patient populations could significantly influence and potentially accelerate its subsequent development pathway.
The CERTIS1 trial (NCT05417594) commenced on June 24, 2022, and is actively recruiting participants, with an estimated primary completion date of February 19, 2027.[9]
The available information predominantly outlines the trial's design, objectives, and the robust preclinical rationale supporting AZD-9574's development. Specific clinical results, such as detailed efficacy outcomes or comprehensive patient safety data from the CERTIS1 trial, are not yet publicly detailed in the provided materials.[8] This is standard for an ongoing clinical trial in its early phases. General statements indicate that it is "too early to know for sure" about the clinical benefits, but researchers are hopeful for outcomes such as tumor shrinkage and improved survival.[8] Regulatory summaries describe CERTIS1 as a first-in-human study primarily designed to evaluate safety and tolerability.[4] Searches for specific clinical trial results presented at major oncology conferences like ASCO or ESMO in 2024 did not yield detailed outcomes for AZD-9574 or CERTIS1 within the provided dataset.[9]
Given AZD-9574's differentiated profile, particularly its PARP1 selectivity and BBB penetration, coupled with the comprehensive and strategically designed CERTIS1 trial, there is considerable anticipation within the oncology community for the initial clinical data readouts. These results, when available, will be critical in determining whether the promising preclinical attributes of AZD-9574 translate into meaningful clinical benefit and a favorable safety profile in patients, especially concerning its activity in CNS malignancies.
The primary goal of the ongoing Phase I/IIa CERTIS1 study is to rigorously assess the safety and tolerability of AZD-9574, both when administered as a monotherapy and in combination with other anti-cancer agents.[4] Clinical trial protocols detail comprehensive safety monitoring, including the systematic collection and grading of all Adverse Events (AEs) and Serious Adverse Events (SAEs). Dose Limiting Toxicities (DLTs) will be carefully evaluated during the dose-escalation phases to establish the maximum tolerated dose (MTD) or recommended Phase II dose (RP2D).[8] Standard safety assessments involve regular monitoring of laboratory tests (hematology, liver and kidney function), electrocardiograms (ECGs), vital signs, and ECOG performance status.[8]
As the CERTIS1 trial is currently ongoing, detailed clinical safety data from patients treated with AZD-9574 are not yet publicly available in the provided information.[3] The emergence of such data will be crucial for understanding the human safety profile of this next-generation PARP inhibitor. Given AZD-9574's designed capacity for significant BBB penetration, clinical safety monitoring will likely include vigilant observation for any potential CNS-specific adverse events. While enhanced CNS exposure is desirable for treating brain tumors, it also means that brain tissue will be exposed to higher concentrations of the drug. Therefore, beyond the systemic toxicities commonly associated with PARP inhibitors (such as gastrointestinal or hematological effects), a critical aspect of the safety evaluation in CERTIS1 will be the careful characterization of any neurological AEs. The therapeutic benefit derived from CNS penetration must be carefully weighed against any potential increase in CNS-related toxicity.
A central hypothesis in the development of AZD-9574 is that its high selectivity for PARP1, with minimal inhibition of PARP2, will translate into a more favorable safety profile compared to first-generation, non-selective PARP inhibitors.[3] This is particularly anticipated with respect to hematologic toxicities, such as anemia, neutropenia, and thrombocytopenia, which can be dose-limiting for older PARP inhibitors.
The preclinical evidence supporting this hypothesis is compelling. Mechanistically, PARP2 inhibition (and subsequent trapping on DNA) has been strongly implicated as a contributor to the hematologic toxicity observed in patients treated with non-selective PARP inhibitors.3 By selectively targeting PARP1, AZD-9574 is designed to avoid these PARP2-mediated effects.
Indeed, preclinical studies have shown that:
If this improved safety profile, especially the reduction in hematologic toxicity, is confirmed in human clinical trials, AZD-9574 could offer a significantly wider therapeutic window. This improved tolerability could allow for more sustained dosing at therapeutically effective levels, better patient compliance, and, importantly, more feasible and tolerable combination regimens with other myelosuppressive therapies. Such an advantage would not only enhance patient quality of life but also fundamentally improve the drug's overall clinical utility and versatility, particularly in complex combination strategies where managing cumulative toxicity is a critical challenge.
AZD-9574 is distinguished by its high selectivity for PARP1, exhibiting over 8000-fold greater affinity for PARP1 compared to PARP2, and it was specifically designed to minimize PARP2 inhibition.[3] This contrasts with first-generation PARP inhibitors such as olaparib, rucaparib, niraparib, and talazoparib, which are generally non-selective and inhibit both PARP1 and PARP2 to varying degrees.[3] The rationale for this targeted selectivity is based on evidence suggesting that PARP1 inhibition is primarily responsible for the desired anti-cancer efficacy in HRD+ cells, while PARP2 inhibition is a significant contributor to dose-limiting hematologic toxicities.[3]
AZD-9574 is part of an evolving landscape in PARP inhibitor development that is increasingly focused on achieving greater target specificity to optimize the risk-benefit profile. Other PARP1-selective inhibitors are also in development, including AZD5305 (another agent from AstraZeneca, though notably not CNS penetrant), saruparib (also from AstraZeneca), NMS-293, and HH102007.[11] The emergence of multiple such agents underscores that PARP1 selectivity is a key area of research and competition, aiming to refine PARP-targeted therapy. This trend reflects a maturation in the field, moving beyond broad PARP inhibition to more precise targeting of PARP1, coupled with effective mechanisms like DNA trapping, while minimizing off-target effects. This sets the stage for potentially more refined, tolerable, and ultimately more effective therapies.
A second major differentiating feature of AZD-9574 is its designed and demonstrated ability to efficiently cross the BBB and achieve significant CNS exposure. Multiple preclinical studies across species (mouse, rat, cynomolgus monkey) have confirmed this, with reported unbound brain-to-plasma partition coefficients (Kpuu) of approximately 0.17 (mouse), 0.31 (rat), and 0.79 (monkey).[3] PET imaging studies in non-human primates provided direct visual confirmation of AZD-9574's brain distribution.[3]
This contrasts significantly with most first-generation PARP inhibitors, which generally exhibit poor or very limited BBB penetration:
While some other PARP inhibitors, such as pamiparib (Kpuu ~0.093; B:P ratio ~0.2) and veliparib (B:P ~0.3-1), have reported BBB penetration, and NMS-293 is suggested to have potentially even higher CNS accumulation (B:P ~4-10) [3], AZD-9574's overall profile is particularly compelling. The combination of high CNS penetration, potent PARP1-selective inhibition, and effective PARP1-DNA trapping [3] positions AZD-9574 as a potentially best-in-class agent for CNS applications. An effective CNS drug requires not only good BBB penetration but also potent target engagement and an acceptable safety profile within the brain. AZD-9574 appears to integrate these critical attributes, underpinning its potential as a leading candidate for CNS malignancies, rather than possessing just one of these features in isolation. It is also important to note that AZD5305, despite its PARP1 selectivity, is not CNS penetrant, further highlighting the unique combination of properties in AZD-9574.[14]
The distinct pharmacological characteristics of AZD-9574—namely its PARP1 selectivity and superior BBB penetration—are anticipated to translate into significant clinical advantages:
The improved CNS penetration and potentially better safety profile of AZD-9574 may also allow for the successful application of PARP inhibition in tumor types or clinical settings where first-generation PARP inhibitors were either ineffective due to poor drug delivery or too toxic to be administered optimally. This is particularly relevant in neuro-oncology and for heavily pre-treated patients who require less toxic therapeutic options. Consequently, AZD-9574 is poised not only to improve upon existing indications for PARP inhibitors but also to potentially expand their utility to new patient populations and clinical scenarios previously considered challenging or unsuitable for this class of drugs.
Table 4 provides a comparative summary of AZD-9574 against select first-generation PARP inhibitors.
Table 4: Comparative Profile of AZD-9574 vs. Select First-Generation PARP Inhibitors
Feature | AZD-9574 | Olaparib | Rucaparib | Niraparib | Talazoparib |
---|---|---|---|---|---|
PARP1 Selectivity | High (>8000x vs PARP2) | Non-selective (PARP1/2) | Non-selective (PARP1/2) | Non-selective (PARP1/2) | Non-selective (PARP1/2) |
PARP2 Inhibition | Minimal / Designed to avoid | Yes | Yes | Yes | Yes |
BBB Penetration (Qualitative; Kpuu or B:P if available) | High (e.g., rat Kpuu ~0.31, monkey Kpuu ~0.79) 3 | Low/Minimal (Kpuu <0.02) 3 | Low/Minimal (ineffective CNS penetration) 16 | Moderate (B:P 0.1-0.2, but minimal PET signal) 3 | Low/Minimal (Kpuu <0.02) 3 |
Primary Mechanism of Hematotoxicity (Hypothesized) | Minimized due to PARP1 selectivity | Linked to PARP2 inhibition | Linked to PARP2 inhibition | Linked to PARP2 inhibition | Linked to PARP2 inhibition |
Key Differentiator(s) | PARP1-selective trapper + High CNS penetration | --- | --- | --- | Potent PARP trapper |
References for table content primarily include:.[3]
The unique pharmacological profile of AZD-9574 positions it with significant therapeutic potential across several key oncological indications, particularly those involving the CNS and tumors with HRR deficiencies.
The compelling combination of PARP1 selectivity, efficient PARP1-DNA trapping, and high CNS penetration underpins the assertion that AZD-9574 has the potential to be a best-in-class PARP inhibitor, especially for the treatment of both primary and secondary brain tumors.[3]
The therapeutic utility of AZD-9574 is being extensively explored in combination strategies, reflecting the broader trend in oncology towards multi-modal treatments.
Due to its targeted mechanism, anticipated improved safety profile stemming from PARP1 selectivity, and unique CNS accessibility, AZD-9574 has the potential to serve as a versatile backbone for combination therapies. This versatility could span a range of cancer types and involve diverse therapeutic partners, including chemotherapy, ADCs, and potentially other targeted agents or immunotherapies, tailored to specific tumor biology and clinical contexts, both within and outside the CNS.
AZD-9574 is positioned as a "next-wave" or "next-generation" PARP inhibitor, designed to address key limitations of earlier agents in this class, particularly their restricted CNS penetration and the toxicities associated with broader PARP family inhibition (especially PARP2).[3] If its promising preclinical profile translates into clinical success, AZD-9574 could offer a vital new treatment option for patients with advanced cancers who have limited alternatives, with a particular focus on those with CNS involvement or HRD+ tumors.[4]
The development of AZD-9574 aligns with AstraZeneca's broader strategic initiatives in oncology, which emphasize innovation in the DDR space and the advancement of ADCs.[10] The competitive landscape includes other investigational PARP1-selective inhibitors, some of which also claim CNS penetration capabilities (e.g., NMS-293, HH102007).[15] However, AZD-9574's specific and well-documented combination of high PARP1 selectivity, effective PARP1-DNA trapping, and robust, multi-species evidence of high CNS penetration is central to its differentiated positioning and therapeutic rationale.[3]
The successful clinical development of AZD-9574 could significantly impact the PARP inhibitor market. It has the potential not only to capture a share of existing markets by offering improved efficacy or safety but, more transformatively, to expand the overall market by rendering CNS malignancies—previously largely inaccessible to or poorly responsive to PARP inhibitor therapy—a viable target class for this mechanism of action. This could redefine the scope and applicability of PARP inhibition in oncology.
AZD-9574 (Palacaparib) represents a thoughtfully engineered, next-generation PARP inhibitor with a pharmacological profile strategically designed to overcome key limitations of its predecessors. Its defining features—high selectivity for PARP1 and robust penetration of the blood-brain barrier—are strongly supported by extensive preclinical data. These characteristics underpin its potential to offer improved therapeutic efficacy, particularly in CNS malignancies such as gliomas and brain metastases, and a more favorable safety profile, especially reduced hematologic toxicity, compared to non-selective PARP inhibitors.
The ongoing Phase I/IIa CERTIS1 trial, with its comprehensive modular design, is poised to provide critical insights into the safety, tolerability, pharmacokinetics, pharmacodynamics, and preliminary efficacy of AZD-9574 across a range of HRD+ solid tumors and in various combination regimens, including with temozolomide and novel antibody-drug conjugates. The preclinical evidence of potent monotherapy activity in HRD+ models, significant CNS anti-tumor effects, and synergistic activity with TMZ (potentially allowing for TMZ dose reduction) is highly encouraging.
If the promising preclinical attributes of AZD-9574 are validated in human clinical trials, it could significantly alter the treatment landscape for several hard-to-treat cancers. It holds the potential to become a cornerstone therapy for selected primary and secondary brain tumors and a valuable new option for systemic HRD+ malignancies, potentially offering a wider therapeutic window and improved quality of life for patients. The continued clinical development of AZD-9574 is eagerly anticipated by the oncology community, as it may expand the reach and refine the application of PARP inhibition in cancer therapy.
Published at: June 9, 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.