Small Molecule
C24H26N4O
1616632-77-9
On August 6, 2025, the United States Food and Drug Administration (FDA) granted accelerated approval to dordaviprone, an event that represents a significant milestone in the field of neuro-oncology.[1] Marketed under the brand name Modeyso and developed by Jazz Pharmaceuticals, dordaviprone is a first-in-class, orally administered small molecule protease activator.[1] This approval establishes dordaviprone as the first and only systemic therapy specifically indicated for the treatment of adult and pediatric patients, aged one year and older, with diffuse midline glioma (DMG) harboring a histone 3 (H3) K27M mutation whose disease has progressed following prior therapy.[1]
The approval of dordaviprone, formerly known by its investigational code ONC201, is the culmination of a developmental journey that began with its discovery by Oncoceutics, followed by acquisition by Chimerix, and ultimately by Jazz Pharmaceuticals.[5] This regulatory decision was met with considerable enthusiasm from the clinical community, with leading experts describing it as a "major turning point in neuro-oncology".[3] For decades, the therapeutic landscape for this devastating central nervous system (CNS) malignancy had remained stagnant, with radiation therapy being the only established standard of care and no approved systemic options available for patients with recurrent or progressive disease.[6] The availability of dordaviprone provides a long-awaited, targeted treatment option for a patient population with a dire prognosis and historically limited therapeutic avenues.[3]
The approval of dordaviprone is not merely a therapeutic advance but also a powerful validation of a biomarker-driven drug development strategy within neuro-oncology, a field that has been characterized by the failure of numerous non-targeted agents. The precise targeting of a specific, well-defined patient population—those with the H3 K27M mutation—was instrumental in demonstrating a clear clinical benefit signal in a rare and aggressive disease. This success was predicated on a deep understanding of the tumor's underlying biology, where the H3 K27M mutation serves as both a prognostic marker and a key oncogenic driver, creating a specific vulnerability that dordaviprone exploits.[3] This approach stands in stark contrast to previous strategies that treated high-grade gliomas as a monolithic entity. Consequently, the success of dordaviprone establishes a new precedent for future drug development in CNS tumors, underscoring the critical importance of molecular characterization at diagnosis to guide therapeutic selection.
Diffuse midline glioma is a rare, aggressive, and uniformly lethal high-grade (WHO Grade IV) glioma that arises in the midline structures of the central nervous system, including the thalamus, brainstem (pons), and spinal cord.[9] A significant portion of these tumors were previously classified as diffuse intrinsic pontine glioma (DIPG) before the 2016 and 2021 WHO classifications of CNS tumors redefined the category based on its molecular signature.[5] The disease predominantly affects children and young adults, with an estimated 2,000 new cases in the U.S. each year.[5] The prognosis is exceptionally poor, with a median overall survival of approximately one year from the time of initial diagnosis and less than six months following disease progression after frontline radiation therapy.[3]
The defining molecular hallmark of this glioma subtype is a specific missense mutation, H3 K27M, which occurs in the genes encoding histone H3 proteins, most commonly H3F3A (encoding H3.3) or HIST1H3B/C (encoding H3.1).[3] This mutation results in the substitution of the amino acid lysine (K) with methionine (M) at position 27 of the histone H3 tail.[8] Histone proteins are critical for packaging DNA into chromatin, and modifications to their tails, such as methylation, regulate gene expression. The lysine at position 27 is a key site for trimethylation (H3K27me3), an epigenetic mark associated with gene silencing that is deposited by the Polycomb Repressive Complex 2 (PRC2).[8]
The H3 K27M mutation acts as a dominant-negative inhibitor of PRC2's methyltransferase activity, leading to a global reduction, or hypomethylation, of H3K27me3 throughout the genome.[8] This widespread epigenetic dysregulation results in an aberrant gene expression profile, promoting the expression of oncogenes and driving tumor proliferation and growth.[3] This specific biological consequence of the H3 K27M mutation is the central vulnerability targeted by dordaviprone, making the presence of the mutation an essential biomarker for treatment eligibility.[8] The approval of dordaviprone thus elevates comprehensive molecular testing for the H3 K27M mutation to an indispensable component of the standard of care for all patients diagnosed with a midline glioma.
Dordaviprone is a first-in-class small molecule belonging to the imipridone chemical class.[12] As an investigational compound, it was widely known as ONC201 and also referred to as TIC10.[14] Its formal approval brings with it a standardized nomenclature and a set of unique identifiers crucial for its precise identification in clinical, regulatory, and research contexts. A comprehensive summary of its chemical and physical properties is provided in Table 1.
The molecule is classified as an organic heterotricyclic compound, with a complex structure defined by its IUPAC name: 11-benzyl-7-[(2-methylphenyl)methyl]-2,5,7,11-tetrazatricyclo[7.4.0.0$^{2,6}$]trideca-1(9),5-dien-8-one.[14] This structure confers its ability to penetrate the blood-brain barrier, a critical property for any agent targeting CNS tumors, and to engage with its intracellular targets.[11]
Table 1: Key Physicochemical and Structural Properties of Dordaviprone
Category | Property | Value / Identifier | Source(s) |
---|---|---|---|
Nomenclature | Generic Name | Dordaviprone | 14 |
Brand Name | Modeyso | 3 | |
Former Names | ONC201, TIC10 | 14 | |
Identifiers | CAS Number | 1616632-77-9 | 1 |
DrugBank ID | DB14844 | 1 | |
PubChem CID | 73777259 | 14 | |
UNII | 9U35A31JAI | 14 | |
Chemical Properties | Molecular Formula | C24H26N4O | 14 |
Molecular Weight | 386.49 g/mol | 15 | |
IUPAC Name | 11-benzyl-7-[(2-methylphenyl)methyl]-2,5,7,11-tetrazatricyclo[7.4.0.0$^{2,6}$]trideca-1(9),5-dien-8-one | 14 | |
Drug Class | Chemical Class | Imipridone | 12 |
Pharmacological Class | Protease Activator, Dopamine D2 Receptor Antagonist | 1 |
Dordaviprone exerts its antineoplastic effects through a unique, first-in-class mechanism of action characterized by its selective and simultaneous engagement with two distinct molecular targets: the G-protein coupled dopamine receptor D2 (DRD2) and the mitochondrial caseinolytic protease P (ClpP).[12] This dual-targeting, or bitopic, activity distinguishes dordaviprone from other targeted therapies and is central to its efficacy in H3 K27M-mutant glioma.[22] While early investigations into its mechanism suggested a primary role as an inhibitor of the Akt and ERK signaling pathways, subsequent direct binding studies have clarified that DRD2 and ClpP are the principal binding partners, with effects on Akt/ERK being downstream consequences of DRD2 antagonism.[15] The identification of ClpP as a direct target was a pivotal discovery that resolved earlier debate and solidified the current understanding of dordaviprone's mode of action.[23]
This dual mechanism represents a sophisticated therapeutic strategy. By concurrently targeting a cell surface receptor involved in proliferation signals (DRD2) and an intracellular mitochondrial enzyme critical for metabolic homeostasis (ClpP), dordaviprone launches a multi-pronged assault on the tumor cell.[12] This approach attacks the cancer through two distinct but complementary angles: first, by disrupting critical pro-survival signaling cascades, and second, by inducing a catastrophic failure of cellular energy production.[11] Such a multi-faceted mechanism may contribute to the durable responses observed in some patients, as it presents a greater challenge for the development of therapeutic resistance compared to agents that act on a single pathway. For a tumor cell to evade dordaviprone's effects, it would need to develop compensatory mechanisms to overcome both the signaling blockade and the metabolic collapse, a statistically less probable event than overcoming a single point of failure.
One of the two core mechanisms of dordaviprone is its function as an antagonist of the dopamine receptor D2 (DRD2).[10] Preclinical studies have revealed that DRD2 is frequently overexpressed in H3 K27M-mutant glioma cells, providing a clear biological rationale for targeting this receptor.[11] By binding to and inhibiting DRD2, dordaviprone effectively blocks the receptor's downstream signaling activity, which plays a role in tumor cell proliferation and survival.[21]
The antagonism of DRD2 leads to the inactivation of key downstream signaling pathways, including the RAS/MAPK and PI3K/Akt pathways.[21] This results in the reduced phosphorylation and activity of critical effector proteins such as extracellular signal-regulated kinase (ERK) and protein kinase B (Akt).[21] The inhibition of these pathways has several antitumor consequences. One significant effect is the activation of the transcription factor FOXO3a. When Akt is active, it phosphorylates FOXO3a, sequestering it in the cytoplasm. Dordaviprone-mediated Akt inhibition allows FOXO3a to translocate to the nucleus, where it transcriptionally upregulates the gene for TNF-related apoptosis-inducing ligand (TRAIL), a potent pro-apoptotic cytokine.[16] The collective result of DRD2 antagonism is the induction of an integrated stress response and the promotion of programmed cell death, or apoptosis, in the tumor cells.[3]
The second, and equally critical, component of dordaviprone's mechanism is its role as an allosteric agonist of the mitochondrial protease ClpP.[3] ClpP is a highly conserved serine protease located within the mitochondrial matrix, where it plays a role in mitochondrial protein quality control.[10] Dordaviprone binds to an allosteric site on the ClpP enzyme, inducing a conformational change that leads to its hyperactivation.[11]
This hyperactivation of ClpP results in the dysregulated and indiscriminate degradation of a wide range of mitochondrial proteins, including essential components of the electron transport chain and enzymes involved in oxidative phosphorylation.[11] This effectively sabotages the cell's primary energy-producing machinery. Tumor cells, particularly aggressive ones like DMG, are often highly reliant on mitochondrial metabolism to meet their high energetic and biosynthetic demands. By disrupting this fundamental process, dordaviprone induces a state of severe metabolic stress, impairs mitochondrial function, and ultimately triggers a TRAIL-independent pathway of apoptosis.[3] The expression level of ClpP itself has been identified as a potential biomarker of response to dordaviprone, further cementing its importance as a key therapeutic target.[10]
The ultimate therapeutic consequence of dordaviprone's dual mechanism is its ability to counteract the core molecular pathology of H3 K27M-mutant glioma. The profound alteration of mitochondrial metabolism induced by ClpP hyperactivation has a direct downstream effect on the cell's epigenetic landscape.[3] Specifically, this metabolic disruption leads to the restoration of H3K27 trimethylation (H3K27me3) in tumor cells that harbor the H3 K27M mutation.[3]
By reversing the pathognomonic loss of this critical epigenetic silencing mark, dordaviprone helps to normalize the aberrant gene expression profile that drives the oncogenic state.[3] This remarkable ability to link a metabolic intervention to the correction of an epigenetic defect represents the convergence of dordaviprone's dual mechanisms of action on the fundamental driver of the disease. It is this integrated effect—disrupting signaling, collapsing metabolism, and restoring epigenetic balance—that underpins its clinical activity. The validation of ClpP as a druggable target in oncology through dordaviprone's success is a significant scientific advance. It opens a new therapeutic avenue for exploration, suggesting that other ClpP agonists could be developed not only for DMG but also for a range of other malignancies that exhibit ClpP upregulation or a pronounced dependency on mitochondrial metabolism.[10] This could herald the emergence of a new class of "mitochondrial-targeted" cancer therapies.
The clinical pharmacology of dordaviprone has been well-characterized, defining its behavior in the human body and informing its dosing regimen and potential for interactions.
Absorption: Dordaviprone is formulated as an oral capsule for once-weekly administration.[3] Following oral intake, it is rapidly absorbed, with the time to reach maximum plasma concentration (
Tmax) occurring at a median of 1.4 hours post-dose in a fasted state.[21] The effect of food on its absorption has been studied; administration with a high-fat meal decreases the peak plasma concentration (
Cmax) by approximately 40% and delays the Tmax to 2.25 hours.[21] However, the total systemic exposure, as measured by the area under the concentration-time curve (AUC), remains unchanged.[21] This indicates that while food slows the rate of absorption, it does not affect the overall amount of drug absorbed. Despite this, the approved labeling recommends administration on an empty stomach (at least one hour before or three hours after a meal).[21] For patients who have difficulty swallowing, the capsule contents may be mixed with certain liquids (e.g., sports drinks, apple juice) or sprinkled on applesauce, with studies showing this method of administration has a minimal impact on total drug exposure.[21]
Distribution: Dordaviprone exhibits extensive distribution into tissues, as evidenced by a large apparent volume of distribution (Vd) of 450 L.[21] It is highly bound to plasma proteins, with a binding fraction of 95-97%.[21] A critical characteristic for a drug targeting a CNS malignancy is its ability to cross the blood-brain barrier. Preclinical and clinical studies have confirmed that dordaviprone is CNS-penetrant, achieving therapeutic concentrations within the tumor tissue, which is essential for its clinical activity.[11]
Metabolism: The primary route of elimination for dordaviprone is hepatic metabolism, mediated predominantly by the cytochrome P450 (CYP) enzyme system.[21] In vitro and clinical studies have identified CYP3A4 as the major enzyme responsible for its metabolism, with minor contributions from CYP2D6, CYP2B6, CYP2C8, CYP2C9, and CYP3A5.[21] The major circulating metabolite identified in human plasma is ONC207, an N-dealkylated product which has been shown to be pharmacologically inactive.[32]
Excretion: Following extensive metabolism, the resulting metabolites of dordaviprone are eliminated from the body primarily through renal excretion. Mass balance studies using radiolabeled dordaviprone showed that approximately 71% of the administered dose is recovered in the urine, with an additional 20% recovered in the feces.[29] The excretion of unchanged, parent dordaviprone is minimal, accounting for less than 0.3% of the total dose in excreta, which confirms that metabolic clearance is the near-exclusive pathway for its elimination.[32] The mean terminal elimination half-life (
t1/2) of dordaviprone is approximately 11 hours.[21]
The pharmacokinetic profile of dordaviprone is characterized by rapid absorption, extensive distribution, and metabolism-dependent clearance. A summary of the key steady-state pharmacokinetic parameters in adults following the recommended dose is provided in Table 2.
Table 2: Summary of Key Pharmacokinetic Parameters of Dordaviprone
Parameter | Value | Description | Source(s) |
---|---|---|---|
Tmax (Median) | 1.4 hours | Time to reach maximum plasma concentration | 21 |
Cmax | 2.8 mcg/mL | Maximum plasma concentration | 21 |
AUC | 23 mcg·hr/mL | Area under the plasma concentration-time curve (total exposure) | 21 |
Volume of Distribution (Vd) | 450 L | Apparent volume of distribution | 21 |
Protein Binding | 95-97% | Fraction of drug bound to plasma proteins | 21 |
Elimination Half-life (t1/2) | 11 hours | Time for plasma concentration to decrease by half | 21 |
Clearance (CL/F) | 27 L/hr | Apparent total clearance from plasma after oral administration | 21 |
The heavy reliance of dordaviprone on CYP3A4 for its metabolic clearance makes it susceptible to clinically significant drug-drug interactions (DDIs).[29] This represents the drug's primary clinical pharmacology liability and necessitates careful management of concomitant medications. Co-administration of dordaviprone with strong inhibitors of CYP3A4 can lead to a substantial increase in dordaviprone plasma concentrations and overall exposure. A clinical DDI study with itraconazole, a potent CYP3A4 inhibitor, demonstrated a 4.5-fold increase in the AUC of dordaviprone.[32] Such an increase in exposure could heighten the risk of concentration-dependent toxicities, such as QTc interval prolongation.[3] Consequently, the prescribing information recommends avoiding concomitant use with strong or moderate CYP3A4 inhibitors. If co-administration is unavoidable, a dose reduction of dordaviprone is required, along with increased patient monitoring.[3]
Conversely, co-administration with drugs that are strong or moderate inducers of CYP3A4 can accelerate the metabolism of dordaviprone, leading to significantly lower plasma concentrations and potentially compromising its therapeutic efficacy.[3] Therefore, the concomitant use of strong or moderate CYP3A4 inducers with dordaviprone should be avoided.[3] Given that many supportive care medications used in neuro-oncology, including certain antiemetics, anticonvulsants (e.g., carbamazepine, phenytoin), and antifungals (e.g., ketoconazole), are potent modulators of CYP3A4, this potential for DDIs is not a theoretical concern but a highly probable clinical scenario. Meticulous review of all concomitant medications by both the prescribing physician and a clinical pharmacist is a critical safety measure for any patient initiating dordaviprone therapy.
Pharmacokinetic analyses from early-phase studies have explored the relationship between drug exposure and clinical response, leading to dose optimization strategies. A key finding from these studies was the comparison between once-weekly and twice-weekly (on consecutive days) dosing regimens.[35] These analyses showed that while a twice-weekly schedule did not result in drug accumulation or a significant increase in peak concentrations (
Cmax), it did lead to a greater total drug exposure over time, as reflected by a higher AUC.[35]
This observation—that overall exposure could be increased without elevating peak concentrations—formed the scientific rationale for investigating a more intensive dosing schedule. The hypothesis is that greater or more sustained target engagement, driven by a higher cumulative AUC, might translate into improved clinical efficacy. This principle is being formally tested in the pivotal Phase 3 ACTION trial, which includes both a once-weekly and a twice-weekly dordaviprone arm, in addition to a placebo arm.[18] The results from this trial will provide definitive evidence on the optimal dosing strategy. If the twice-weekly arm demonstrates superior efficacy, it would suggest that for this particular mechanism of action, sustained pressure on the tumor's metabolic and epigenetic systems is a more critical determinant of outcome than intermittent high-level disruption. This finding could have broader implications, informing the development of other novel agents that target similar biological pathways.
The accelerated approval of dordaviprone by the FDA was not based on a single, large, randomized controlled trial, but rather on a robust, pre-specified integrated efficacy analysis of a pooled population from five separate open-label, non-randomized clinical studies conducted in the United States.[1] This approach is often utilized for rare diseases with high unmet need where conducting large randomized trials is challenging. The trials included in this integrated analysis were ONC006 (NCT02525692), ONC013 (NCT03295396), ONC014 (NCT03416530), ONC016 (NCT05392374), and ONC018 (NCT03134131).[1]
The efficacy population consisted of 50 adult and pediatric patients who had received single-agent dordaviprone for recurrent H3 K27M-mutant DMG.[1] Key eligibility criteria for inclusion in this analysis were stringent, requiring patients to have histologically confirmed H3 K27M-mutant diffuse midline glioma with progressive and measurable disease according to the Response Assessment in Neuro-Oncology-High Grade Glioma (RANO-HGG) criteria.[1] Patients were required to be at least 90 days post-completion of radiation therapy, have had an adequate washout period from any prior anticancer therapies, and possess a Karnofsky Performance Status (KPS) or Lansky Performance Status (LPS) score of ≥60, indicating a reasonable level of functional independence.[1] Patients with primary spinal tumors, diffuse intrinsic pontine glioma, or cerebrospinal fluid dissemination were excluded from this specific efficacy analysis.[1]
The primary efficacy outcome for the integrated analysis was the Overall Response Rate (ORR), which was rigorously assessed by a blinded independent central review (BICR) using the updated RANO 2.0 criteria.[1] The analysis demonstrated a statistically significant and clinically meaningful ORR of 22%, with a 95% confidence interval of 12% to 36%.[1] This means that approximately one in five patients in this heavily pre-treated population experienced a significant reduction in tumor size.
While a 22% ORR may appear modest in some oncologic settings, its significance must be interpreted within the context of the disease. For recurrent H3 K27M-mutant DMG, for which there were no approved therapies and a median survival of less than six months post-progression, achieving objective tumor responses is a substantial clinical achievement.[3] The true value of these responses is further underscored by their durability. The key secondary endpoint, Duration of Response (DOR), revealed that these responses were not transient. The median DOR was 10.3 months (95% CI: 7.3 to 15.2 months).[1] This durability is arguably the most compelling aspect of the efficacy data. A durable response lasting nearly a year for a patient expected to survive less than six months represents a profound clinical benefit, transforming the therapeutic goal from short-term palliation to the possibility of meaningful, long-term disease control for this subset of responding patients. Further analysis of the 11 patients who achieved an objective response showed that 73% maintained their response for at least six months, and 27% maintained their response for at least twelve months.[1]
Table 3: Summary of Pivotal Efficacy Data from Integrated Analysis
Endpoint | Value (95% Confidence Interval) | Source(s) |
---|---|---|
Overall Response Rate (ORR) | 22% (12% - 36%) | 1 |
Median Duration of Response (DOR) | 10.3 months (7.3 - 15.2 months) | 1 |
Responders with DOR ≥ 6 months | 73% | 1 |
Responders with DOR ≥ 12 months | 27% | 1 |
Disease Control Rate (DCR) | 40% (26% - 55%) | 11 |
The clinical benefit of dordaviprone extended beyond radiographic tumor shrinkage, encompassing improvements in patient-centric outcomes that directly impact quality of life. A significant challenge in managing patients with high-grade gliomas is the reliance on high-dose corticosteroids (like dexamethasone) to control neurological symptoms and peritumoral edema. Long-term steroid use is associated with severe and debilitating side effects, including myopathy, hyperglycemia, immunosuppression, and Cushing's syndrome. In the integrated analysis, among evaluable patients who were on corticosteroids at baseline, 46.7% were able to achieve a sustained reduction of 50% or more in their daily corticosteroid dose while maintaining stable or improved neurological function.[4] This steroid-sparing effect is a crucial clinical benefit, reducing treatment-related morbidity for patients.
Furthermore, dordaviprone treatment was associated with improvements in neurological function. Among 34 evaluable patients, 20.6% experienced a confirmed improvement in their KPS or LPS performance score.[4] An improvement in performance status signifies an enhancement in a patient's ability to carry out daily activities and reflects a tangible improvement in their overall well-being and neurological health. These data highlight the importance of measuring outcomes beyond tumor response in neuro-oncology. The ability to reduce steroid dependence and improve functional status are highly meaningful benefits for patients and their families. These findings suggest that future clinical trials in this field should consider incorporating such patient-centric endpoints as key secondary or even co-primary measures of a drug's overall clinical value.
The safety and tolerability of dordaviprone were evaluated in a pooled population of 376 adult and pediatric patients with glioma who were enrolled in four open-label clinical studies.[3] Overall, dordaviprone was found to be well-tolerated. In the pivotal efficacy cohort of 50 patients, the tolerability profile was particularly favorable, with no Grade 4 treatment-related treatment-emergent adverse events (TEAEs) reported. Furthermore, there were no treatment-related deaths or discontinuations due to drug toxicity in this cohort.[4]
This favorable tolerability is a critical attribute of the drug. The median time to response with dordaviprone can be prolonged, on the order of several months (e.g., a median of 8.3 months in one analysis).[12] A drug with a high burden of cumulative or severe toxicity might force patients to discontinue treatment before they have an adequate opportunity to respond. The manageable safety profile of dordaviprone allows for the sustained, long-term administration necessary for patients to potentially derive the durable clinical benefits observed in the efficacy analyses.
Common Adverse Reactions: The most frequently reported TEAEs (occurring in ≥20% of patients) were generally low-grade and consistent with symptoms often experienced by patients with CNS tumors. These included fatigue (34%), headache (32%), vomiting (24%), nausea (24%), and musculoskeletal pain (20%).[3] For pediatric patients specifically, nausea and vomiting were noted as common adverse events.[28]
Serious Adverse Reactions: Serious adverse reactions (SARs) were reported in 33% of patients in the broader safety population.[3] The SARs reported in more than 2% of patients included hydrocephalus (fluid buildup in the brain, 5%), vomiting (4.3%), headache (3.2%), seizure (2.4%), and muscular weakness (2.1%).[3] A persistent challenge in neuro-oncology clinical trials is the difficulty in definitively attributing such neurological events to either drug toxicity or the natural progression of the underlying brain tumor. The placebo-controlled data from the ongoing ACTION trial will be essential to provide a clearer delineation of the drug's true safety profile versus the background event rate in this patient population. Fatal adverse reactions occurred in 1% of patients, which included cardiac arrest, intracranial hemorrhage, and encephalopathy.[3]
Laboratory Abnormalities: The most common Grade 3 or 4 laboratory abnormalities (occurring in ≥2% of patients) included decreased lymphocyte count (7%), decreased calcium (2.7%), and increased alanine aminotransferase (ALT) (2.4%).[3] Routine blood monitoring is recommended to track these parameters.[28]
The prescribing information for dordaviprone includes three significant warnings and precautions that require careful clinical management.[1]
Dordaviprone has been shown to cause concentration-dependent prolongation of the QTc interval on an electrocardiogram (ECG), which is a known risk factor for potentially life-threatening ventricular tachyarrhythmias such as torsades de pointes.[3] In the clinical trial safety population, a QTcF increase of >60 msec from baseline occurred in 6% of patients, and a QTcF value >500 msec was observed in 1.2% of patients.[3] To mitigate this risk, specific monitoring is required. An ECG and serum electrolytes should be assessed prior to initiating therapy and then periodically throughout treatment as clinically indicated. The frequency of this monitoring should be increased for patients with pre-existing risk factors, such as congenital long QT syndrome, a history of ventricular arrhythmias, electrolyte abnormalities, or heart failure. Importantly, because dordaviprone exposure is significantly increased by CYP3A4 inhibitors, patients receiving these concomitant medications also require more frequent monitoring. Concomitant use of dordaviprone with other drugs known to prolong the QT interval should be avoided if possible. If unavoidable, increased monitoring and separation of administration times are recommended. Dose interruption, reduction, or permanent discontinuation may be necessary if significant QTc prolongation develops.[3]
Severe hypersensitivity reactions, including anaphylaxis, can occur with dordaviprone. In the pooled safety population, Grade 3 hypersensitivity reactions were reported in 0.3% of patients.[3] Clinicians and patients must be aware of the signs and symptoms, which may include rash, hives, fever, hypotension, wheezing, or angioedema (swelling of the face or throat). Patients should be instructed to seek immediate medical attention if such symptoms occur. If a clinically significant hypersensitivity reaction occurs, dordaviprone should be immediately interrupted and appropriate medical treatment initiated. Depending on the severity of the reaction, temporary interruption or permanent discontinuation of the drug may be warranted.[3]
Based on its mechanism of action and findings from animal studies, dordaviprone can cause fetal harm when administered to a pregnant woman.[3] Therefore, it is contraindicated for use during pregnancy. All patients of reproductive potential must be advised of this risk. Females of reproductive potential should be advised to use effective contraception during treatment with dordaviprone and for at least one month after the final dose. Similarly, male patients with female partners of reproductive potential should be advised to use effective contraception during treatment and for one month following the last dose to prevent fetal exposure.[3]
The recommended dosage of dordaviprone is administered orally once weekly, on the same day each week.[3]
For adult patients and pediatric patients weighing at least 52.5 kg, the recommended dose is 625 mg once weekly.[28]
For pediatric patients weighing less than 52.5 kg, the dosage is scaled according to body weight to achieve comparable exposures to the adult population. The specific weight-based dosing recommendations are detailed in Table 4. Dosing has not been established for pediatric patients weighing less than 10 kg.[3]
Table 4: Recommended Dosing for Pediatric Patients by Body Weight
Body Weight Range | Recommended Once-Weekly Dose | Source(s) |
---|---|---|
10 kg to <12.5 kg | 125 mg | 41 |
12.5 kg to <27.5 kg | 250 mg | 41 |
27.5 kg to <42.5 kg | 375 mg | 41 |
42.5 kg to <52.5 kg | 500 mg | 41 |
≥52.5 kg | 625 mg | 41 |
Dordaviprone should be taken on an empty stomach, either at least one hour before or three hours after food intake.[21] The capsules should be swallowed whole. For patients who are unable to swallow capsules, the contents can be opened and mixed with 15-30 mL of a sports drink, apple juice, lemonade, or water and administered orally or via a feeding tube immediately or within two hours of preparation.[21]
Dordaviprone's path to market was facilitated by several of the FDA's expedited programs, which are designed to accelerate the development and review of drugs that treat serious conditions and fill an unmet medical need.[1] The New Drug Application (NDA) was submitted by Chimerix in December 2024 and was subsequently accepted for review by the FDA in February 2025.[9] Recognizing the drug's potential to provide a significant improvement in the treatment of a serious disease, the FDA granted the application Priority Review, which shortens the target review timeline.[9]
Throughout its development, dordaviprone had also received Fast Track designation, Orphan Drug designation for glioma, and Rare Pediatric Disease designation.[1] These designations provide various incentives, including more frequent interactions with the FDA during development (Fast Track), market exclusivity and financial incentives (Orphan Drug), and a potential Priority Review Voucher upon approval (Rare Pediatric Disease). The review process was further streamlined by the applicant's use of the Assessment Aid, a voluntary submission that helps organize the application data to facilitate a more efficient FDA assessment.[1] This concerted use of regulatory tools culminated in the accelerated approval on August 6, 2025, nearly two weeks ahead of its scheduled PDUFA date.[1]
The accelerated approval of dordaviprone was based on the surrogate endpoint of overall response rate. As a condition of this approval, the sponsor is required to conduct a confirmatory clinical trial to verify and describe the drug's clinical benefit, typically by demonstrating an improvement in a clinical endpoint such as overall survival (OS) or progression-free survival (PFS).[5] This requirement is being fulfilled by the ongoing ACTION trial (NCT05580562).[9]
The ACTION trial is a large, international, randomized, double-blind, placebo-controlled Phase 3 study with an estimated enrollment of 450 patients.[45] Its design represents a strategic move to evaluate dordaviprone in an earlier treatment setting to maximize its potential clinical benefit. The trial enrolls patients with newly diagnosed H3 K27M-mutant diffuse glioma who have completed standard frontline radiotherapy but have not yet experienced disease progression.[9] This adjuvant-like setting, when tumor burden is theoretically at its lowest, is often where therapeutic agents can have their greatest impact on long-term outcomes.
Eligible patients are randomized in a 1:1:1 ratio to one of three treatment arms:
The co-primary efficacy endpoints of the ACTION trial are Overall Survival (OS) and Progression-Free Survival (PFS), as assessed by BICR using RANO-HGG criteria.[22] Key secondary endpoints include safety and tolerability, quality of life measures, and clinical benefit markers such as corticosteroid response.[18] The outcome of this trial is paramount; a positive result will be required to convert the accelerated approval to a full, traditional approval and will definitively establish dordaviprone's role in the frontline management of H3 K27M-mutant DMG.[5] A successful outcome would fundamentally alter the standard of care for these patients, shifting it from "radiation followed by watchful waiting" to "radiation followed by adjuvant dordaviprone."
As of late 2025, dordaviprone's marketing authorization is limited to the United States. There is currently no active application for marketing authorization submitted to the European Medicines Agency (EMA) or the United Kingdom's Medicines and Healthcare products Regulatory Agency (MHRA).[48] This creates a significant disparity in access for patients outside the U.S.
For patients in Europe and other regions where dordaviprone is not yet approved, access may be possible through several alternative pathways. One option is enrollment in an ongoing clinical trial, such as the ACTION study, which has sites in 17 countries.[48] Other potential avenues include compassionate use or managed access programs, where applicable, or importation for personal use under "named patient" regulations, which exist in many countries.[48] However, these pathways can be complex and are not universally available. This global access disparity highlights a growing challenge in the era of accelerated approvals for biomarker-defined rare cancers. The success of dordaviprone in the U.S. will likely serve as a key case study in ongoing discussions among international regulatory bodies about developing more harmonized and agile review pathways to ensure that patients with rare and life-threatening diseases have equitable and timely access to transformative new therapies.
The approval of dordaviprone for recurrent DMG marks the beginning, not the end, of its clinical investigation. The most immediate next step is the completion of the ACTION trial to establish its role in the frontline setting.[7] Beyond this, several other avenues of research are being considered by experts in the field.
One area of interest is the development of dordaviprone-based combination therapies. Combining dordaviprone with other agents, potentially including other targeted therapies or immunotherapies, could lead to synergistic effects and improved outcomes.[7] Another area for exploration is the use of dordaviprone in other clinical contexts. This includes its potential use in patients with H3 K27M-mutant gliomas that arise in non-midline locations of the brain, a rare subset of tumors where early case reports suggest potential activity.[7] Furthermore, research is needed to identify other tumor types that may harbor the H3 K27M mutation and could therefore be sensitive to dordaviprone.[7] Preclinical data have also suggested potential antitumor activity of dordaviprone in other malignancies where its targets are overexpressed, such as endometrial cancer, lung cancer, and glioblastoma without the H3 K27M mutation, though these investigations remain highly exploratory.[24]
The accelerated approval of dordaviprone (Modeyso) is a watershed moment for the neuro-oncology community and, most importantly, for patients and families affected by H3 K27M-mutant diffuse midline glioma. As the first systemic therapy to demonstrate meaningful and durable clinical benefit in this devastating disease, it addresses a critical unmet medical need that has persisted for decades.
Dordaviprone's novel, dual-targeting mechanism of action—simultaneously antagonizing the DRD2 receptor and hyperactivating the mitochondrial protease ClpP—represents a sophisticated and multi-pronged attack on the tumor's fundamental biology. This mechanism not only disrupts pro-survival signaling and induces metabolic catastrophe but also leads to the restoration of the normal epigenetic state, directly counteracting the primary oncogenic driver of the disease.
The clinical data, derived from a carefully integrated analysis of five studies, demonstrated an objective response rate of 22% with a remarkable median duration of response exceeding 10 months. Coupled with a manageable safety profile that permits long-term administration, these results provide a new therapeutic option that can offer a significant extension of high-quality life for a subset of patients.
The approval of dordaviprone serves as a powerful testament to the value of a precision medicine approach in neuro-oncology. Its success will undoubtedly catalyze further research into targeted therapies for other molecularly defined subsets of brain tumors and places a new, urgent emphasis on comprehensive molecular testing as a standard of care at diagnosis. While the results of the confirmatory Phase 3 ACTION trial are awaited to solidify its role in the frontline setting, dordaviprone has already fundamentally changed the treatment paradigm and provided a tangible source of hope for a community that has long awaited a breakthrough.
Published at: September 7, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.