Small Molecule
C29H39NO3
96346-61-1
Onapristone (DrugBank ID: DB12637) is an investigational, orally bioavailable, synthetic steroidal small molecule classified as a Type I, or "pure," competitive antagonist of the progesterone receptor (PR). First described in 1984, its initial development for breast cancer and as an endometrial contraceptive showed significant promise. However, this was abruptly halted during Phase III trials in 1995 due to an unacceptable incidence of severe hepatotoxicity associated with the immediate-release (IR) formulation.
The compound was later revived based on the pharmacokinetic hypothesis that the liver toxicity was driven by high peak plasma concentrations ($C_{max}$). This led to the development of an extended-release (ER) formulation, ONA-XR, designed to lower $C_{max}$ while maintaining therapeutic drug exposure. Recent clinical investigations with ONA-XR have successfully demonstrated a markedly improved safety profile, with no definitive cases of drug-induced liver injury meeting Hy's Law criteria.
Despite this pharmaceutical success, the clinical efficacy of ONA-XR has been modest. While a Phase 0 study in early breast cancer (ONAWA) confirmed its antiproliferative biological activity, a Phase II trial in heavily pre-treated metastatic breast cancer (SMILE) failed to demonstrate any objective responses. More promising signals have emerged from combination therapy, particularly in the Phase II OATH trial, where Onapristone plus anastrozole showed encouraging activity in heavily pre-treated endometrial cancer.
Pharmacokinetically, Onapristone is characterized by high plasma protein binding (99.5%) and metabolism primarily via cytochrome P450 3A4 (CYP3A4), an enzyme it also inactivates, creating a high potential for drug-drug interactions. As of April 2023, the most recent developer, Context Therapeutics, ceased internal development of ONA-XR and is exploring strategic options, effectively halting its progression. Onapristone remains an unmarketed investigational agent. Its history serves as a compelling case study on the ability of formulation science to overcome critical safety issues, but also underscores the formidable challenge of demonstrating compelling clinical efficacy in the modern oncology landscape, even with a mechanistically sound and safer compound.
The trajectory of Onapristone's development is a multi-decade narrative characterized by initial scientific promise, a critical safety failure, and a subsequent, scientifically driven revival that ultimately faced new challenges. This history provides crucial context for its current status and future potential.
Onapristone was first developed by Schering and described in the scientific literature in 1984.[1] As a potent and highly selective antiprogestin, it was initially investigated for clinical use in hormone-sensitive conditions, most notably for the treatment of breast cancer and as a potential endometrial contraceptive.[1] Early clinical studies with an immediate-release (IR) formulation were encouraging, demonstrating clear anti-tumor activity and establishing proof-of-concept in breast cancer patients.[2]
However, the program was abruptly terminated during Phase III clinical trials in 1995. This decision was precipitated by the discovery that a majority of patients treated with the IR formulation developed significant liver function abnormalities.[1] The hepatotoxicity was considered severe enough to render the risk-benefit profile unacceptable, leading to the cessation of all development activities and shelving of the compound for over a decade.
Years after its discontinuation, a re-evaluation of Onapristone's pharmacology and pharmacokinetic (PK) data led to a new hypothesis regarding the cause of the observed hepatotoxicity. The prevailing theory was that the liver injury was not an intrinsic property of the molecule itself but rather an off-target effect related to the very high peak plasma concentrations ($C_{max}$) achieved with the IR formulation.[2] This hypothesis provided a clear scientific rationale for reviving the drug: if the $C_{max}$ could be reduced while maintaining a therapeutically effective total drug exposure (Area Under the Curve, or AUC), the drug's therapeutic index might be dramatically improved.
This led to a deliberate pharmaceutical reformulation program. The result was the creation of an extended-release (ER) oral tablet, known as ONA-XR, designed specifically to slow the rate of drug absorption. The intended PK profile of ONA-XR was a blunted $C_{max}$ and a more sustained plasma concentration over a longer period, allowing for effective twice-daily (BID) dosing and continuous suppression of the progesterone receptor while avoiding the sharp peaks associated with the original IR formulation.[6]
With the new ER formulation in hand, Onapristone re-emerged in the clinical landscape under the stewardship of companies including Arno Therapeutics and, more recently, Context Therapeutics.[12] A new wave of clinical trials was initiated to evaluate ONA-XR in a variety of progesterone receptor-positive (PR+) cancers, including prostate, endometrial, breast, and ovarian cancers.[1]
This modern development phase successfully validated the core hypothesis of the reformulation strategy, demonstrating a significantly improved safety profile, particularly with respect to hepatotoxicity. However, the clinical efficacy results were mixed, and the program encountered challenges with slow patient accrual in some trials.[15] In a decisive turn of events, Context Therapeutics announced in April 2023 that it was ceasing internal development of ONA-XR and was "exploring strategic options" for the asset.[15] This decision, coupled with the termination of associated clinical trials, has effectively archived the program, leaving Onapristone as an unmarketed investigational agent with an uncertain future. The drug's history illustrates a key principle in modern drug development: mitigating a critical safety flaw, while a significant scientific achievement, does not guarantee clinical or commercial success if the efficacy profile is not sufficiently compelling to compete in a crowded and demanding therapeutic landscape.
Onapristone is a synthetic, steroidal small molecule whose chemical structure and properties are fundamental to its biological activity and pharmacokinetic behavior.
Onapristone is unambiguously identified by a standardized set of chemical and regulatory codes. Its IUPAC name is (8S,11R,13R,14S,17S)-11-[4-(dimethylamino)phenyl]-17-hydroxy-17-(3-hydroxypropyl)-13-methyl-1,2,6,7,8,11,12,14,15,16-decahydrocyclopenta[a]phenanthren-3-one.[14] Over its long development history, it has been referred to by numerous synonyms and developmental codes, including ZK-98299, ZK-299, IVV-1001, AR-18, and, for its extended-release formulation, ONA-XR and Apristor.[1] Key identifiers are consolidated in Table 1.
Onapristone's chemical formula is $C_{29}H_{39}NO_{3}$, corresponding to a molecular weight of approximately 449.6 g/mol.[1] It belongs to the class of organic compounds known as oxosteroids, which are steroid derivatives containing a carbonyl group.[18] Its steroidal backbone provides a rigid scaffold for interaction with nuclear hormone receptors.
Key properties influencing its absorption, distribution, metabolism, and excretion (ADME) profile include its very low aqueous solubility (0.00759 mg/mL) and high lipophilicity, as indicated by a logP value between 3.96 and 4.63.[18] Despite its low solubility, predictive models suggest it has a high bioavailability of 1, indicating efficient absorption from the gastrointestinal tract.[18] The molecule contains two hydrogen bond donors and four hydrogen bond acceptors, contributing to its receptor-binding interactions.[17]
The distinction between Onapristone's two primary oral formulations is central to its developmental story.
Table 1: Key Identifiers and Physicochemical Properties of Onapristone
| Parameter | Value | Source(s) |
|---|---|---|
| DrugBank ID | DB12637 | 16 |
| CAS Number | 96346-61-1 | 1 |
| UNII | H6H7G23O3N | 16 |
| Molecular Formula | $C_{29}H_{39}NO_{3}$ | 1 |
| Molecular Weight | 449.635 g/mol (Average) | 1 |
| IUPAC Name | (8S,11R,13R,14S,17S)-11-[4-(dimethylamino)phenyl]-17-hydroxy-17-(3-hydroxypropyl)-13-methyl-1,2,6,7,8,11,12,14,15,16-decahydrocyclopenta[a]phenanthren-3-one | 14 |
| InChIKey | IEXUMDBQLIVNHZ-YOUGDJEHSA-N | 14 |
| Water Solubility | 0.00759 mg/mL | 18 |
| logP | 3.96 - 4.63 | 18 |
| Chemical Class | Steroid; Oxosteroid | 1 |
Onapristone's therapeutic rationale is rooted in its specific and potent mechanism as a progesterone receptor antagonist. Its pharmacological profile distinguishes it from other agents in its class and has been validated in numerous preclinical cancer models.
Onapristone is a Type I, or "pure," competitive antagonist of the progesterone receptor (PR).[1] This classification as a "silent" antagonist means that it binds to the PR but is incapable of activating transcription, thereby fully antagonizing the functions of progesterone.[22]
At the molecular level, its mechanism of action is multifaceted. Upon binding to the PR, Onapristone induces a conformational change that prevents the formation of functional PRA and PRB dimers. This action inhibits both ligand-dependent (progesterone-induced) and ligand-independent (e.g., growth factor-induced) phosphorylation of the receptor, a key step in its activation. A specific site of action is the inhibition of phosphorylation at serine 294 (PR-S294) and the prevention of the nuclear translocation of this activated form of the receptor.[14] By blocking these critical activation steps, Onapristone prevents the PR from associating with its co-activators and, crucially, from binding to progesterone response elements on DNA. This complete blockade of PR-mediated gene transcription is the foundation of its potential antineoplastic activity in hormone-driven cancers that overexpress the PR.[8]
Onapristone demonstrates high-affinity binding to the progesterone receptor, with reported dissociation constants ($K_d$) of 1.6 nM and 11.9 nM for human endometrium and myometrium PR, respectively, confirming its high potency.[25] While its primary target is the PR, it also possesses some additional, weaker hormonal activities. It exhibits reduced antiglucocorticoid activity and minimal antiandrogenic activity, making it more selective for the PR compared to other antiprogestins.[1]
A comparison with the archetypal antiprogestin, mifepristone (RU486), highlights Onapristone's distinct pharmacological profile. The most significant difference lies in their interaction with the PR. While Onapristone is a pure or silent antagonist, mifepristone is a weak partial agonist.[1] This means that while mifepristone generally blocks progesterone action, in certain cellular contexts it can weakly activate the receptor, potentially leading to mixed or unpredictable effects. Onapristone's pure antagonist nature was theorized to provide a more complete and reliable blockade of PR signaling. Furthermore, Onapristone is estimated to be 10- to 30-fold more potent as an antiprogestogen than mifepristone and has a more favorable selectivity profile with its reduced off-target antiglucocorticoid activity.[1] This theoretically superior mechanism formed the core scientific rationale for its development as a next-generation antiprogestin. However, the modest clinical efficacy observed in later trials raises important questions about whether this "cleaner" mechanism translates to superior clinical outcomes, or if this mode of PR antagonism is simply insufficient to drive deep responses in advanced cancers.
Table 2: Pharmacological Comparison of Onapristone and Mifepristone
| Feature | Onapristone | Mifepristone (RU486) | Source(s) |
|---|---|---|---|
| Mechanism on PR | Pure ("Silent") Antagonist | Partial Agonist / Antagonist | 1 |
| Antiprogestogen Potency | 10- to 30-fold higher | Baseline | 1 |
| Antiglucocorticoid Activity | Reduced | Significant | 1 |
| Antiandrogenic Activity | Little / Minimal | Present | 1 |
| Clinical Status | Never Marketed | Approved for medical termination of pregnancy | 1 |
Extensive preclinical research has validated Onapristone's anti-cancer activity and supported its clinical development. Studies have consistently shown that it is active in multiple preclinical models of hormone-dependent cancers.[27] It has demonstrated tumor-inhibitory effects in animal models, including rat and mouse mammary tumors, with efficacy comparable to that of tamoxifen or oophorectomy.[21] In vitro studies on breast cancer cell lines have confirmed its ability to reduce cell viability and induce apoptosis.[25]
More recent preclinical work has focused on its potential in combination therapies. In patient-derived xenograft (PDX) models of ER+/PR+ breast cancer, combining Onapristone with standard-of-care agents such as the selective estrogen receptor degrader (SERD) fulvestrant, the CDK4/6 inhibitor palbociclib, or the PI3K inhibitor alpelisib resulted in significantly increased anti-tumor activity compared to monotherapy. A triple-therapy combination led to tumor regression in a majority of the xenografts, providing a strong rationale for clinical trials evaluating these combinations.[30] Other preclinical studies have suggested that progesterone can promote tumor growth by decreasing immune cell infiltration, providing a basis for investigating antiprogestins like Onapristone to promote an anti-tumor immune response.[30]
The absorption, distribution, metabolism, and excretion (ADME) profile of Onapristone is critical to understanding its efficacy, safety, and potential for drug-drug interactions. Its high lipophilicity, extensive metabolism, and very high protein binding are defining features.
Onapristone is orally bioavailable and well-absorbed from the gastrointestinal tract.[16] The formulation significantly impacts its absorption kinetics. A clinical study of the 10 mg IR formulation in healthy female subjects found that administration with a high-fat, high-calorie meal delayed the time to maximum plasma concentration ($T_{max}$) from 1 hour in the fasted state to 4 hours in the fed state. This was accompanied by a statistically significant decrease in $C_{max}$ of approximately 18% and a small increase in total exposure ($AUC_{0-\infty}$) of approximately 13%.[32] As these changes were within acceptable regulatory limits, it was concluded that Onapristone could be administered without regard to food.[32] The ER formulation was specifically engineered to have a reduced rate of absorption compared to the IR form, resulting in a delayed $T_{max}$ (2.5 hours for ER vs. 1 hour for IR) and a significantly lower day 1 $C_{max}$.[34]
Following absorption, Onapristone undergoes extensive distribution into tissues. A key characteristic of its distribution is its exceptionally high binding to plasma proteins, reported to be 99.5% in human plasma.[31] This high degree of protein binding means that only a very small fraction (0.5%) of the drug in circulation is unbound, or "free," to exert its pharmacological effect, be metabolized, or be excreted. This property makes the drug highly susceptible to displacement interactions. If a co-administered drug displaces even a small amount of Onapristone from its binding proteins, it can cause a disproportionately large, and potentially toxic, increase in the free drug concentration. For example, a shift in binding from 99.5% to 99.0% would double the free concentration from 0.5% to 1.0%, a 100% increase in the active fraction. This mechanism may have been an underappreciated contributor to the unpredictable toxicity observed in early trials.
Onapristone is primarily metabolized in the liver. In vitro studies using human liver microsomes have conclusively identified cytochrome P450 3A4 (CYP3A4) as the principal enzyme responsible for its N-demethylation, a primary metabolic pathway.[35] The main metabolite produced is N-mono-desmethyl onapristone, also known as M1.[32]
Complicating its metabolic profile, Onapristone is not only a substrate of CYP3A4 but also a mechanism-based, time-dependent inactivator of the enzyme.[36] This dual role means that Onapristone can inhibit its own metabolism over time and has a high potential to interfere with the metabolism of a wide range of other drugs that are also CYP3A4 substrates. This complex interaction with a major drug-metabolizing enzyme is a critical consideration for potential drug-drug interactions.
Detailed human mass balance studies quantifying the routes and proportions of Onapristone excretion are not available in the provided materials. One secondary source indicates that the drug and its metabolites are excreted via both urine and feces, which is typical for lipophilic, hepatically metabolized compounds.[37] The inclusion of renal function criteria (e.g., creatinine clearance < 40-60 mL/min) as an exclusion criterion in multiple clinical trial protocols suggests that renal clearance plays a role in the elimination of Onapristone or its metabolites, and that impairment could lead to drug accumulation.[8]
The pharmacokinetic profile of Onapristone is heavily dependent on the formulation and dosing regimen.
Across the tested dose range of the ER formulation (10 mg to 50 mg BID), both the AUC and $C_{max}$ have been shown to be dose-proportional, indicating predictable pharmacokinetics within this therapeutic window.[7]
Table 3: Summary of Key Pharmacokinetic Parameters (IR vs. ER Formulations)
| Parameter | 100 mg IR QD | 50 mg ER BID | Source(s) |
|---|---|---|---|
| $T_{max}$ (Day 1) | ~1 hour | ~2.5 hours | 32 |
| $C_{max}$ (Day 1) | Significantly higher | >2-fold lower than IR | 34 |
| Half-life ($t_{1/2}$) | ~2-4 hours (single dose) | 8-12 hours (repeated dosing) | 7 |
| Time to Steady State | Not reported | ~200 hours | 7 |
Onapristone has been evaluated in a range of clinical trials across several PR-positive cancer types. Its clinical journey reflects a transition from promising monotherapy in early studies to a more nuanced role in combination therapies and biomarker-selected populations in its recent development phase.
Clinical development has focused on malignancies where the progesterone receptor is believed to be a driver of tumor growth, including breast, endometrial, ovarian, and prostate cancers.[14] The program's evolution saw a shift from the original IR formulation to the current ER formulation, with trials spanning Phase 0 to Phase II.
Onapristone has also been investigated in various stages of prostate cancer, including androgen-independent, recurrent, and metastatic disease, based on the expression of PR in these tumors.[16] As of 2016, prostate cancer was a primary focus for its redevelopment.[1] However, specific efficacy data from these clinical trials are not detailed in the available documentation.
Table 4: Overview of Major Clinical Trials for Onapristone ER
| Trial ID (Name) | Phase | Indication | Treatment Arm(s) | Key Outcomes | Source(s) |
|---|---|---|---|---|---|
| NCT04142892 (ONAWA) | 0 | Early-stage HR+/HER2- Breast Cancer | Onapristone ER (neoadjuvant) | Met primary endpoint: significant antiproliferative effect (Ki67 reduction). | 40 |
| NCT04738292 (SMILE) | II | Metastatic ER+/HER2- Breast Cancer (post-CDK4/6i) | Onapristone ER + Fulvestrant | Did not meet primary endpoint: 0% ORR in first 11 patients. Median TTP of 63 days. | 42 |
| NCT02052128 | I/II | PR-expressing solid tumors (Breast, Endometrial, Ovarian) | Onapristone ER (dose escalation) | Established RP2D of 50 mg BID. Clinical benefit in 9/52 patients. Good tolerability. | 19 |
| NCT04719273 (OATH) | II | Refractory HR+ Endometrial Cancer | Onapristone ER + Anastrozole | Promising preliminary data: 15.4% ORR, 63.5% 4-month PFS rate, 84.4% DCR. | 44 |
| NCT03909152 | II | Recurrent PR+ Gynecologic Cancers (inc. GCT) | Onapristone ER | No objective responses. In GCT cohort: 35.7% CBR, 20.1% 12-month PFS rate. | 46 |
The safety profile of Onapristone is defined by the historical hepatotoxicity of its IR formulation and the subsequent mitigation of this risk with the ER formulation. The overall safety database for ONA-XR suggests it is a generally well-tolerated agent.
The development of the original IR formulation of Onapristone was halted in 1995 due to an unacceptable rate of liver function test (LFT) abnormalities.[1] The primary goal of the ER formulation was to address this liability.
Clinical data from the modern development program strongly suggest this goal was achieved. A pooled safety analysis of 88 patients from two Phase I-II trials of ONA-XR found that while elevations in alanine aminotransferase (ALT) or aspartate aminotransferase (AST) occurred, their incidence was strongly associated with the presence of liver metastases (20% in patients with liver metastases vs. 6.3% in those without).[2] Of five patients who experienced Grade 3 or higher ALT elevations, four were assessed as unrelated to ONA-XR by a safety data review committee. The fifth case was ultimately adjudicated by an independent hepatologist as unlikely to be drug-related.[48] Critically, across the entire ONA-XR clinical program, there have been no reported cases of drug-induced liver injury meeting Hy's Law criteria (a standard indicator of severe, potentially fatal hepatotoxicity).[5] This indicates that the reformulation strategy was successful in mitigating the primary safety concern.
The overall safety profile of ONA-XR is generally favorable. In a Phase I study, the most common treatment-related adverse events (AEs) were asthenia (21%), increased AST/ALT (17%), and increased gamma-glutamyl transferase (GGT) (10%).[7] Most Grade 3 or higher AEs, particularly LFT elevations, were associated with patients who had progressive disease in the liver.[7] In the OATH trial of ONA-XR plus anastrozole, the most common AEs were mainly Grade 1 or 2 and included hot flashes (36%), increased ALT/AST (29%), nausea (29%), and diarrhea (21%).[44] Across multiple trials, ONA-XR has been described as well-tolerated, with no treatment-related deaths reported.[20]
Table 5: Summary of Common Adverse Events Reported in Onapristone ER Clinical Trials
| Adverse Event | Frequency (%) | Grade | Relevant Trial(s) | Source(s) |
|---|---|---|---|---|
| Asthenia | 21% | Any | Phase I/II (NCT02052128) | 7 |
| AST/ALT Increased | 17-29% | Any | Phase I/II, OATH | 7 |
| GGT Increased | 10% | Grade $\ge$3 | Phase I/II (NCT02052128) | 7 |
| Hot Flashes | 36% | Any | OATH (NCT04719273) | 44 |
| Nausea | 29% | Any | OATH (NCT04719273) | 44 |
| Diarrhea | 21% | Any | OATH (NCT04719273) | 44 |
| Abdominal Pain | N/A | Grade 3 | Phase II (NCT03909152) | 47 |
Based on its mechanism of action and data from clinical trial protocols, several contraindications and precautions are warranted for Onapristone:
Drug interaction databases have identified specific pharmacodynamic risks associated with Onapristone. There is a potential for an increased risk of methemoglobinemia when Onapristone is combined with a wide range of local anesthetics (e.g., benzocaine, lidocaine, procaine) and other compounds like capsaicin and diphenhydramine.[18] Additionally, there is a potential for an increased risk of thrombosis when it is co-administered with erythropoiesis-stimulating agents such as darbepoetin alfa and erythropoietin.[18] These risks warrant careful consideration of concomitant medications.
Onapristone's metabolic profile and high plasma protein binding create a significant potential for clinically relevant drug-drug interactions (DDIs). These interactions can be pharmacokinetic, affecting drug exposure, or pharmacodynamic, altering drug effects.
Onapristone's relationship with the cytochrome P450 3A4 (CYP3A4) enzyme is complex and represents the primary source of pharmacokinetic DDIs. CYP3A4 is responsible for metabolizing approximately half of all drugs on the market, making interactions involving this enzyme particularly common and important.[54]
Pharmacodynamic interactions occur when drugs have additive or antagonistic effects at the receptor or system level. For Onapristone, two key risks have been identified:
Onapristone's very high plasma protein binding of 99.5% presents a significant theoretical risk for displacement interactions.[31] When two highly protein-bound drugs are co-administered, they can compete for binding sites on plasma proteins like albumin. If a second drug displaces Onapristone from these sites, the concentration of the pharmacologically active "free" fraction of Onapristone can increase dramatically, even if the total plasma concentration remains unchanged. This could lead to unexpected toxicity. Caution would be required when co-administering Onapristone with other highly bound drugs, such as warfarin, phenytoin, or certain nonsteroidal anti-inflammatory drugs (NSAIDs).
Despite a long and complex development history spanning several decades and multiple pharmaceutical companies, Onapristone remains an investigational agent that has not achieved regulatory approval in any market.
Onapristone has never been marketed for any clinical indication. It has not received approval from major global regulatory agencies, including the United States Food and Drug Administration (FDA), the European Medicines Agency (EMA), or the Australian Therapeutic Goods Administration (TGA).[1] Records indicate no planned submission for FDA approval, and there is no evidence of a formal review process ever being completed by the EMA or TGA.[12] The drug's development was initially halted in 1995, prior to the EMA's formal establishment, and its subsequent redevelopment has not progressed to the point of a marketing authorization application in any major jurisdiction.
Onapristone is classified as an investigational small molecule.[18] Its most recent chapter of development concluded in April 2023, when Context Therapeutics, the company leading its clinical evaluation, announced the cessation of internal development for the ONA-XR program. The company stated it would be "exploring strategic options" for the asset, a decision often indicative of deprioritization or out-licensing attempts.[15] This halt was attributed to factors including slow patient accrual in key clinical trials, which made it highly unlikely that the program could support a regulatory filing within a viable timeframe.[15] The subsequent termination and withdrawal of related clinical trials, such as the ELONA study (NCT05618613), further confirmed the discontinuation of active development.[12] Consequently, Onapristone is currently an inactive, late-stage clinical asset without a clear path forward to commercialization.
The comprehensive review of Onapristone reveals a drug with a compelling scientific rationale but a challenging and ultimately unsuccessful clinical development path. Its story offers valuable lessons on the interplay between pharmacology, pharmaceutical science, clinical efficacy, and commercial viability in modern oncology.
Onapristone's journey is a cautionary tale. It began as a promising, mechanistically elegant compound—a pure PR antagonist theoretically superior to its predecessors. This promise was cut short by a severe safety signal (hepatotoxicity) that was, at the time, insurmountable. The drug's revival represents a triumph of pharmaceutical science, where a rational, hypothesis-driven reformulation successfully mitigated the primary toxicity, allowing for re-entry into the clinic. However, this second chapter concluded not with a safety failure, but with the inability to demonstrate a sufficiently compelling efficacy profile to justify continued investment in a highly competitive therapeutic area. This arc highlights that while safety is a prerequisite, it does not guarantee success.
The development of ONA-XR stands as a model case for applying pharmacokinetic principles to solve a clinical problem. By identifying high $C_{max}$ as the likely driver of toxicity and engineering a formulation to specifically address it, the developers successfully improved the drug's therapeutic index. The clinical data for ONA-XR clearly show a vastly improved hepatic safety profile compared to the historical data for the IR formulation. This achievement was necessary for the drug to have any chance of progression. However, it was not sufficient. The modest efficacy results, particularly the lack of objective responses in the SMILE trial for metastatic breast cancer, suggest that the level of clinical activity, even with a safer formulation, fell short of the high bar required for new oncology agents, especially in heavily pre-treated patient populations.
Given the recent halt in development, the future of Onapristone is highly uncertain and contingent on a new sponsor acquiring the asset with a clear and focused strategy. Based on the available data, several potential niches could be explored to maximize its chances of success:
Onapristone is a pharmacologically sophisticated molecule and a testament to the power of formulation science to rescue a promising compound from a critical safety flaw. However, its journey also serves as a stark reminder that in the landscape of modern oncology, a sound mechanism of action and an acceptable safety profile are necessary but not sufficient conditions for success. The inability to translate its biological activity into compelling clinical responses in advanced disease has, for a second time, halted its path to the clinic. Without a new champion and a refined clinical strategy focused on rational combinations, biomarker selection, and appropriate disease settings, Onapristone is likely to remain a well-documented but unfulfilled chapter in the history of endocrine cancer therapy.
Published at: October 29, 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.