C33H44F2N2O3
1474034-05-3
Friedreich's Ataxia
Omaveloxolone, marketed under the brand name SKYCLARYS®, represents a landmark therapeutic advancement in the field of neurodegenerative diseases. It is the first and only medication approved by major global regulatory bodies, including the U.S. Food and Drug Administration (FDA) and the European Commission (EC), for the treatment of Friedreich's ataxia (FA).[1] This approval is indicated for adults and adolescents aged 16 years and older who are living with this rare, inherited, and relentlessly progressive condition.[3] FA is characterized by debilitating neurological and systemic symptoms, including ataxia, muscle weakness, loss of coordination, cardiomyopathy, and a significantly shortened lifespan, for which no disease-modifying therapies previously existed.[4] The advent of Omaveloxolone signals a paradigm shift, moving the management of FA beyond purely supportive and symptomatic care to an era of active pharmacological intervention designed to slow the underlying progression of the disease.[7] Its approval is a culmination of years of collaborative effort between patients, researchers, advocacy organizations, and industry, offering tangible hope to a community that has long faced a profound unmet medical need.[7]
From a chemical standpoint, Omaveloxolone (also identified by the internal development code RTA-408) is a novel, semi-synthetic small molecule drug.[1] It belongs to the oleanane triterpenoid class of compounds, which are derived from natural plant-based precursors and have been subjected to extensive chemical modification to optimize their therapeutic properties.[1] Specifically, Omaveloxolone is a second-generation synthetic oleanane triterpenoid, engineered to be a potent activator of the Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signaling pathway, a master regulator of cellular antioxidant and anti-inflammatory responses.[4] Its complex pentacyclic structure, which includes organofluorine and nitrile functional groups, distinguishes it as a highly specialized agent designed to engage specific molecular targets implicated in the pathophysiology of FA.[1]
The path to regulatory approval for Omaveloxolone was facilitated by its recognition as a potential breakthrough for a serious and rare condition. The drug was developed by Reata Pharmaceuticals, which successfully navigated the regulatory landscape by securing multiple expedited designations from the FDA. These included Orphan Drug, Fast Track, Priority Review, and Rare Pediatric Disease designations, each reflecting the urgency and high unmet need for an effective FA treatment.[5] This concerted effort culminated in its landmark approval by the FDA on February 28, 2023.[4] Following its success in the United States, the European Commission granted marketing authorization in February 2024, extending its availability to patients within the European Union.[1] In a significant corporate development, Reata Pharmaceuticals was acquired by the global biotechnology company Biogen in September 2023.[13] This acquisition was strategically positioned to leverage Biogen's extensive global commercial and medical infrastructure to accelerate the worldwide delivery and accessibility of SKYCLARYS® to the global FA patient community.[13]
Property | Value | Source(s) |
---|---|---|
Generic Name | Omaveloxolone | 9 |
Brand Name | SKYCLARYS® | 2 |
Synonyms/Internal Codes | RTA-408, RTA 408 | 1 |
DrugBank ID | DB12513 | 4 |
CAS Number | 1474034-05-3 | 4 |
UNII | G69Z98951Q | 1 |
Molecular Formula | C33H44F2N2O3 | 1 |
Molecular Weight | 554.723 g/mol | 4 |
IUPAC Name | N-((4aS,6aR,6bS,8aR,12aS,14aR,14bS)-11-cyano-2,2,6a,6b,9,9,12a-heptamethyl-10,14-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,12a,14,14a,14b-octadecahydropicen-4a-yl)-2,2-difluoropropanamide | 1 |
Chemical Class | Semi-synthetic Oleanane Triterpenoid, Pentacyclic Triterpenoid, Organofluorine Compound | 1 |
Physical State | Crystalline solid | 15 |
Solubility | Practically insoluble in water | 1 |
pKa | 7.26 | 1 |
To comprehend the therapeutic action of Omaveloxolone, it is essential to first understand the molecular pathology of Friedreich's ataxia. FA is an autosomal recessive neurodegenerative disorder caused by a genetic mutation in the frataxin (FXN) gene.[5] Specifically, an unstable expansion of a GAA trinucleotide repeat sequence within the first intron of the
FXN gene leads to epigenetic modifications and transcriptional silencing, resulting in a profound deficiency of the mitochondrial protein frataxin.[16] Frataxin plays a critical role in mitochondrial iron homeostasis and the biosynthesis of iron-sulfur clusters, which are essential cofactors for numerous enzymes involved in the electron transport chain and cellular metabolism.[10]
The resulting frataxin deficiency triggers a cascade of downstream pathological events. The primary consequence is severe mitochondrial dysfunction, characterized by impaired adenosine triphosphate (ATP) production, leading to a cellular energy crisis.[10] This is compounded by the accumulation of iron within the mitochondria, which catalyzes the formation of highly damaging reactive oxygen species (ROS) via the Fenton reaction.[16] The combination of energy failure and excessive ROS generation creates a state of chronic oxidative stress, which, along with concurrent neuroinflammation, drives the progressive degeneration of neurons in the dorsal root ganglia, spinal cord, and cerebellum, as well as damage to cardiomyocytes and pancreatic beta cells.[10] A critical feature of this pathological state is the functional impairment of the Nrf2 pathway, the body's primary defense mechanism against oxidative stress. In cells from FA patients and in animal models of the disease, Nrf2 expression and activity are suppressed, leaving cells vulnerable to the ongoing oxidative damage.[11]
The central mechanism of action of Omaveloxolone is the potent pharmacological activation of the Nrf2 transcription factor, directly addressing the pathway's suppression in FA.[1] This mechanism is far more sophisticated than that of conventional antioxidant supplements. Instead of acting as a simple scavenger of free radicals, Omaveloxolone functions as a molecular switch that amplifies the body's own comprehensive and coordinated antioxidant defense system.[4]
Under normal physiological conditions, the activity of Nrf2 is tightly regulated by its principal negative regulator, Kelch-like ECH-associated protein 1 (KEAP1).[9] KEAP1 acts as an adaptor protein that binds to Nrf2 in the cytoplasm, targeting it for ubiquitination and subsequent proteasomal degradation, thereby keeping Nrf2 levels low.[20] Omaveloxolone is designed to disrupt this interaction. It functions as an electrophilic small molecule that forms a reversible covalent bond with a specific, highly reactive cysteine residue (Cys151) on the KEAP1 protein.[20] This molecular modification induces a conformational change in KEAP1, inhibiting its ability to bind and degrade Nrf2.[9]
By preventing its degradation, Omaveloxolone allows newly synthesized Nrf2 to escape KEAP1's control, accumulate in the cytoplasm, and translocate into the cell nucleus.[9] Once inside the nucleus, Nrf2 dimerizes with small Maf proteins and binds to specific DNA sequences known as Antioxidant Response Elements (AREs), which are located in the promoter regions of hundreds of cytoprotective genes.[20] This binding event initiates the coordinated transcription and subsequent translation of a vast network of proteins that collectively restore cellular homeostasis.
The Nrf2-mediated gene expression program triggered by Omaveloxolone leads to a multi-pronged therapeutic effect that directly counteracts the core pathologies of FA.
A primary consequence of Nrf2 activation is the robust upregulation of a wide array of antioxidant enzymes.[20] These include phase II detoxification enzymes like NAD(P)H quinone:oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1), as well as proteins crucial for the synthesis and regeneration of glutathione (GSH), the most abundant endogenous antioxidant.[10] Furthermore, Nrf2 boosts the production of NADPH, a critical reducing equivalent required by many of these antioxidant enzymes to function.[10] By orchestrating this broad-spectrum antioxidant response, Omaveloxolone effectively quenches the excessive ROS produced due to mitochondrial dysfunction, mitigates oxidative damage to lipids, proteins, and DNA, and restores cellular redox balance.[11] This approach is fundamentally different from and superior to the administration of exogenous antioxidants like Vitamin E or Coenzyme Q10. While those agents provide a finite, stoichiometric effect, Omaveloxolone acts catalytically, amplifying the entire endogenous protective network for a more powerful and sustained therapeutic outcome.[4]
Beyond its antioxidant role, the therapeutic action of Omaveloxolone is enhanced by its ability to suppress inflammation. The Nrf2 pathway has a well-established reciprocal inhibitory relationship with the pro-inflammatory Nuclear Factor-kappa B (NF-κB) signaling pathway.[4] By activating Nrf2, Omaveloxolone effectively dampens NF-κB activity, leading to reduced production of pro-inflammatory cytokines and chemokines.[20] This dual anti-oxidative and anti-inflammatory activity is particularly relevant in FA, where neuroinflammation is a key driver of neuronal cell death and disease progression.[17]
Omaveloxolone also directly addresses the root cause of the energy deficit in FA by improving mitochondrial health and function.[20] The reduction of oxidative stress within the mitochondria protects sensitive components of the electron transport chain from damage, preserving their efficiency. Furthermore, Nrf2 activation upregulates genes involved in mitochondrial biogenesis, fatty acid oxidation, and overall metabolic efficiency.[4] This restoration of mitochondrial bioenergetics leads to increased production of ATP, helping to resolve the cellular energy crisis and support the function of high-energy-demand cells like neurons and cardiomyocytes.[10]
The drug's observed safety profile, particularly the warnings regarding hepatotoxicity and dyslipidemia, can be understood as a direct consequence of its potent and systemic mechanism of action. Nrf2 is a master regulator not only of antioxidant response but also of numerous metabolic and detoxification pathways that are highly active in the liver. The powerful, pharmacological activation of this system by Omaveloxolone can lead to a significant shift in hepatic metabolism. This may manifest as transient cellular stress, reflected by the release of aminotransferases (ALT and AST), as the liver adapts to the new state of heightened metabolic activity. Similarly, Nrf2 plays a complex role in regulating lipid and cholesterol metabolism. Its potent activation can perturb the finely balanced homeostatic control of these pathways, leading to the observed increases in LDL-C and decreases in HDL-C. Therefore, these adverse effects are not idiosyncratic or off-target but are mechanistically coherent with the drug's intended pharmacological action, providing a clear rationale for the specific monitoring protocols required during treatment.
The clinical pharmacology of Omaveloxolone describes its absorption, distribution, metabolism, and excretion (ADME) properties, which are critical for determining the appropriate dosing regimen and understanding its potential for drug interactions.
Omaveloxolone is formulated for oral administration.[4] Following a single dose in the fasted state, it is absorbed with a median time to reach peak plasma concentration (Tmax) of 7 to 14 hours.[22] The total systemic exposure, as measured by the area under the concentration-time curve (AUC), increases in a dose-proportional manner between doses of 50 mg and 150 mg.[9]
A critical aspect of Omaveloxolone's absorption profile is a significant food effect. Administration with a high-fat meal (800-1000 calories) results in a dramatic alteration of its pharmacokinetics. While the total drug exposure (AUC) is only modestly increased by approximately 15%, the peak plasma concentration (Cmax) is increased by a substantial 350%.[9] This disproportionate spike in Cmax underscores the rationale for the strict dosing instruction to administer the drug on an empty stomach. High peak drug concentrations are often associated with an increased risk of concentration-dependent adverse events, particularly hepatotoxicity. By ensuring administration in a fasted state, a more predictable and controlled absorption profile is maintained, minimizing the potential for toxicity associated with high peak levels. This makes patient adherence to the "empty stomach" mandate not merely a recommendation for optimal efficacy but a crucial safety measure to mitigate risk.[5]
Omaveloxolone exhibits extensive distribution throughout the body. It has a very large mean apparent volume of distribution (Vd) of approximately 7,361 L, which indicates that the drug partitions extensively from the plasma into tissues.[9] This is consistent with its lipophilic nature. In the bloodstream, Omaveloxolone is highly bound to plasma proteins, with a binding fraction of 97%.[9]
The metabolism of Omaveloxolone is primarily mediated by the cytochrome P450 (CYP) enzyme system. The major metabolic pathway is through CYP3A4, with minor contributions from CYP2C8 and CYP2J2.[9] This heavy reliance on CYP3A4 for its clearance makes Omaveloxolone susceptible to significant drug-drug interactions with inhibitors or inducers of this enzyme. In addition to being a substrate, Omaveloxolone itself acts as a weak inducer of CYP3A4 and CYP2C8, which can potentially affect the metabolism of other co-administered drugs that are substrates for these enzymes.[1]
Omaveloxolone is eliminated from the body primarily through the fecal route. Following a single radiolabeled oral dose, approximately 92% of the dose was recovered in the feces, with a negligible amount (0.1%) found in the urine, indicating that renal clearance is not a significant pathway for elimination.[22] The drug has a long mean terminal elimination half-life (
t1/2) of approximately 57 hours, which supports the convenient once-daily dosing regimen.[9]
A strong link between drug exposure and pharmacological effect has been established in preclinical models. Studies in non-human primates demonstrated that oral administration of Omaveloxolone resulted in dose-linear plasma pharmacokinetics and dose-dependent induction of Nrf2 target genes in both peripheral blood cells and key tissues like the liver, lung, and brain.[10] Crucially, the systemic plasma exposures that produced significant Nrf2 activation in these animal models were found to be readily achievable and consistent with the exposures observed in FA patients receiving the clinical dose of 150 mg daily.[10] This provides a robust pharmacodynamic bridge, confirming that the clinical dosing regimen achieves exposures sufficient to engage the target pathway and elicit the intended biological response.
The clinical efficacy of Omaveloxolone was definitively established in the pivotal MOXIe (Mitochondrial Ocular Ataxia) trial, a multi-part study that provided the core evidence for its regulatory approvals worldwide.
The approvals by both the FDA and the EMA were primarily based on the robust efficacy and safety data generated from the MOXIe Part 2 study.[4] This phase of the trial was designed as a 48-week, multicenter, randomized, double-blind, placebo-controlled study to rigorously evaluate the effect of Omaveloxolone on disease progression in patients with FA.[6]
The trial enrolled 103 individuals with a genetically confirmed diagnosis of FA, aged between 16 and 40 years, across clinical sites in the United States, Europe, and Australia.[4] Participants were randomized in a 1:1 ratio to receive either Omaveloxolone at a dose of 150 mg once daily or a matching placebo for the 48-week treatment period.[6] A key feature of the study design was the pre-specification of a primary analysis population, known as the Full Analysis Set (FAS), which excluded patients with severe pes cavus (n=82).[31] Pes cavus, a condition of high-arched feet common in FA, can interfere with the assessment of certain gait and stability measures, and its exclusion was intended to reduce variability and enhance the precision of the primary endpoint analysis.[31]
The primary efficacy endpoint of the MOXIe Part 2 trial was the change from baseline in the modified Friedreich's Ataxia Rating Scale (mFARS) score at Week 48.[6] The mFARS is a validated and widely accepted clinical assessment tool specifically designed to quantify the neurological impairment and progression in FA.[33] It is a comprehensive examination that evaluates four key functional domains affected by the disease: bulbar function (swallowing and speech), upper limb coordination, lower limb coordination, and upright stability.[4] Scores range from 0 to 93, with higher scores indicating greater impairment. A decrease in the mFARS score signifies an improvement in neurological function.[31]
The trial met its primary endpoint, demonstrating a statistically significant and clinically meaningful treatment benefit for Omaveloxolone compared to placebo. At the end of the 48-week period, patients in the placebo group experienced the expected disease progression, with their mean mFARS score worsening by +0.85 points from baseline. In stark contrast, patients treated with Omaveloxolone showed a mean improvement in their neurological function, with their mFARS score decreasing by -1.56 points.[17] This resulted in a placebo-corrected least squares mean treatment difference of
-2.41 points (95% Confidence Interval: -4.32 to -0.51; p=0.0138).[28]
The clinical importance of this result is profound. Natural history studies have shown that untreated individuals with FA typically progress, or worsen, on the mFARS scale by approximately 1 to 2 points per year.[30] The -2.41 point difference observed in the MOXIe trial therefore does not represent a mere slowing of progression; it signifies a treatment effect equivalent to halting and reversing approximately one to two years of typical disease progression over the 48-week study period. For patients with a relentlessly degenerative condition, this outcome is highly significant and represents a substantial change in the disease trajectory.[7]
Parameter | SKYCLARYS (n=40) | Placebo (n=42) | Treatment Difference [95% CI] |
---|---|---|---|
Baseline mFARS Score (Mean) | 40.95 | 38.78 | N/A |
LS Mean Change from Baseline at Week 48 | -1.56 | +0.85 | N/A |
Placebo-Corrected Difference | N/A | N/A | -2.41 [-4.32, -0.51] |
p-value | N/A | N/A | 0.0138 |
Source(s): 28 |
Further analysis of the mFARS data revealed that the treatment benefit was consistent across all four functional domains of the scale, with each component numerically favoring Omaveloxolone over placebo.[12] The most pronounced effects were observed in the subscales for upper limb coordination and upright stability, which are directly linked to patients' ability to perform activities of daily living and maintain ambulation.[32] These findings were supported by trends favoring Omaveloxolone in various secondary endpoints, including the Patient Global Impression of Change (PGIC), further reinforcing the overall positive treatment effect.[17]
Given the chronic nature of FA, establishing the durability of a treatment's effect is paramount. Evidence for the long-term benefit of Omaveloxolone comes from two key analyses.
Upon completing the 48-week double-blind phase, all eligible participants were given the option to enroll in an open-label extension (OLE) study, in which all patients received active Omaveloxolone treatment.[4] This design allowed for a "delayed-start" analysis, comparing the long-term trajectory of patients who were on the drug from the beginning (Omav-Omav group) with those who initially received placebo and then switched to the active drug (Placebo-Omav group).[34] The results of this analysis were compelling: the statistically significant difference in mFARS scores that was established at the end of the 48-week placebo-controlled period was maintained and preserved throughout an additional 72 weeks of open-label treatment.[34] This preservation of the initial treatment benefit strongly suggests that Omaveloxolone has a persistent, disease-modifying effect on the course of FA, rather than a purely transient symptomatic one.[34]
To further contextualize the long-term outcomes, a post-hoc, propensity-matched analysis was conducted. This sophisticated analysis compared the 3-year disease progression of patients treated with Omaveloxolone in the MOXIe OLE to a carefully matched control group of untreated patients from a large, prospective natural history study known as the Friedreich's Ataxia Clinical Outcome Measures Study (FA-COMS).[6] The results of this comparison provided powerful supportive evidence for a long-term benefit. Over a 3-year period, the matched untreated FA-COMS cohort progressed by a mean of 6.6 points on the mFARS scale. In contrast, the Omaveloxolone-treated patients from the MOXIe extension progressed by only 3.0 points.[33] This represents a meaningful slowing of disease progression over a multi-year timeframe, reinforcing the conclusion that Omaveloxolone confers a durable and clinically relevant therapeutic benefit.[33]
A comprehensive assessment of the safety and tolerability of Omaveloxolone is essential for its appropriate clinical use. The primary safety data are derived from the pivotal MOXIe trial and its open-label extension.
In the 48-week placebo-controlled portion of the MOXIe trial, Omaveloxolone was generally found to be safe and well-tolerated.[35] While all participants in both the active treatment and placebo groups reported at least one treatment-emergent adverse event (TEAE), the vast majority of these events were classified as mild or moderate in severity.[35] The rate of discontinuation due to adverse reactions was low and comparable between groups, with four patients (8%) in the SKYCLARYS arm and two patients (4%) in the placebo arm discontinuing treatment.[26] The TEAEs that were reported more frequently with Omaveloxolone tended to have their onset within the first 12 weeks of treatment and often decreased in frequency over time, suggesting a degree of adaptation to the medication.[35]
The most frequently reported adverse reactions, defined as those occurring in at least 20% of patients treated with SKYCLARYS and at a higher incidence than placebo, were elevated liver enzymes (aspartate aminotransferase/alanine aminotransferase), headache, nausea, abdominal pain, fatigue, diarrhea, and musculoskeletal pain.[2]
Adverse Reaction | SKYCLARYS (N=51) % | Placebo (N=52) % |
---|---|---|
Elevated Liver Enzymes (AST/ALT) | 37 | 2 |
Headache | 37 | 25 |
Nausea | 33 | 13 |
Abdominal Pain | 29 | 6 |
Fatigue | 24 | 14 |
Diarrhea | 20 | 10 |
Musculoskeletal Pain | 20 | 15 |
Oropharyngeal Pain | 18 | 6 |
Influenza | 16 | 6 |
Vomiting | 16 | 12 |
Muscle Spasms | 14 | 6 |
Back Pain | 13 | 8 |
Decreased Appetite | 12 | 4 |
Rash | 10 | 4 |
Source(s): 29 |
The prescribing information for SKYCLARYS highlights several important warnings and precautions that require careful clinical monitoring and management. These are not unexpected off-target effects but are largely consistent with the drug's potent, systemic biological activity.
The most prominent safety concern associated with Omaveloxolone is the potential for hepatotoxicity, manifested as elevations in serum aminotransferases.[22] In the MOXIe trial, a notable proportion of patients treated with SKYCLARYS experienced increases in ALT and/or AST. Specifically, 31% of patients had elevations greater than three times the upper limit of normal (>3x ULN), and 16% had elevations greater than five times the upper limit of normal (>5x ULN), compared to a near-zero incidence in the placebo group.[22]
Importantly, these transaminase elevations were typically asymptomatic, transient, and reversible upon interruption or discontinuation of the drug.[22] The maximum increases generally occurred within the first 12 weeks of initiating treatment.[22] Critically, there were no observed cases of concomitant elevations in total bilirubin that would meet the criteria for Hy's Law, which is a strong predictor of severe drug-induced liver injury.[22] This suggests that while hepatocellular stress occurs, it does not typically progress to severe, life-threatening liver failure. Nevertheless, this risk necessitates a strict monitoring protocol.
Treatment with Omaveloxolone can lead to an increase in B-type natriuretic peptide (BNP), a biomarker released by the heart in response to ventricular stretch and pressure overload, often used to diagnose and monitor heart failure.[19] In the MOXIe trial, 14% of patients in the SKYCLARYS group experienced a BNP elevation above the upper limit of normal (>100 pg/mL), compared to only 4% of patients in the placebo group.[22] While elevations in BNP can be an indicator of cardiac stress, dedicated cardiac safety assessments conducted during the trial did not reveal any concerning signals. There were no clinically significant differences between the treatment and placebo groups in electrocardiogram (ECG) parameters, echocardiographic findings, or the incidence of cardiac adverse events.[37] This suggests that the observed BNP elevations may not directly correlate with overt cardiac dysfunction in this patient population, but the signal warrants cautious monitoring for any clinical signs or symptoms of fluid overload or heart failure.[25]
Omaveloxolone treatment is also associated with alterations in lipid profiles.[2] Specifically, it can cause an increase in low-density lipoprotein cholesterol (LDL-C, often termed "bad" cholesterol) and a decrease in high-density lipoprotein cholesterol (HDL-C, or "good" cholesterol).[22] In the pivotal trial, the mean increase in LDL-C from baseline at 48 weeks was 23.5 mg/dL, while the mean decrease in HDL-C was 5.3 mg/dL.[22] These changes were observed to occur within the first few weeks of treatment and were reversible, returning to baseline within four weeks of discontinuing the medication.[22] This effect requires periodic monitoring of lipid panels and management according to standard clinical guidelines.[25]
According to the approved prescribing information from regulatory agencies, there are no absolute contraindications to the use of Omaveloxolone.[22]
The safe and effective use of Omaveloxolone requires strict adherence to the prescribing guidelines regarding dosage, administration, patient monitoring, and management of drug interactions.
Due to the identified risks, a specific monitoring plan is a mandatory part of clinical management.
Omaveloxolone's metabolism via CYP3A4 and its own weak inducing effects on CYP3A4 and CYP2C8 create a significant potential for drug-drug interactions that must be carefully managed.
Interacting Drug Class | Example Agents | Effect on Omaveloxolone / Other Drugs | Management Recommendation |
---|---|---|---|
Strong CYP3A4 Inhibitors | Itraconazole, Ketoconazole, Clarithromycin, Ritonavir | Increases Omaveloxolone exposure approximately 4-fold, increasing risk of toxicity. | Avoid concomitant use. If unavoidable, reduce Omaveloxolone dose to 50 mg once daily with close monitoring for adverse reactions. Discontinue co-administration if adverse reactions emerge. |
Moderate CYP3A4 Inhibitors | Verapamil, Diltiazem, Erythromycin, Grapefruit/Grapefruit Juice | Increases Omaveloxolone exposure, increasing risk of toxicity. | Avoid concomitant use. If unavoidable, reduce Omaveloxolone dose to 100 mg once daily. Consider further reduction to 50 mg if adverse reactions emerge. Patients must be advised to avoid grapefruit and grapefruit juice. |
Strong/Moderate CYP3A4 Inducers | Rifampin, Carbamazepine, Phenytoin, St. John's Wort | Significantly decreases Omaveloxolone exposure, which may lead to a loss of therapeutic efficacy. | Avoid concomitant use. |
Hormonal Contraceptives | Estrogen/Progestin-containing products (pills, patches, rings), implants, progestin-only pills | Omaveloxolone is a weak CYP3A4 inducer and may decrease the efficacy of hormonal contraceptives, leading to risk of unintended pregnancy. | Avoid concomitant use. Advise patients of childbearing potential to use an effective non-hormonal contraceptive method (e.g., intrauterine system, barrier methods) during treatment and for 28 days after the final dose. |
Sensitive CYP3A4/CYP2C8 Substrates | Midazolam (CYP3A4), Repaglinide (CYP2C8) | Omaveloxolone may decrease the plasma concentrations and efficacy of these co-administered drugs. | Monitor for lack of efficacy of the co-administered substrate. Refer to the prescribing information of the concomitant medication for potential dose adjustments. |
Source(s): 2 |
Omaveloxolone (SKYCLARYS®) is a first-in-class, oral, once-daily Nrf2 activator that constitutes a historic breakthrough in the therapeutic landscape for Friedreich's ataxia.[4] By targeting the fundamental pathophysiological mechanisms of the disease—mitochondrial dysfunction, oxidative stress, and neuroinflammation—it has demonstrated a statistically significant and clinically meaningful capacity to slow the relentless neurological progression of FA.[28] The approval of Omaveloxolone transforms the standard of care, moving it from a paradigm of purely supportive management to one of active, disease-modifying intervention. It provides the first validated pharmacological tool to alter the natural history of FA, offering new hope and a tangible therapeutic option to patients, families, and clinicians who have long awaited such an advance.[7]
The clinical utility of Omaveloxolone is defined by a careful balance of its benefits against its known risks. The primary benefit—slowing the progression of a severe, life-shortening neurodegenerative disorder—is substantial. This must be weighed against its principal safety concerns, which include the potential for transient, reversible hepatotoxicity, elevations in the cardiac biomarker BNP, and unfavorable changes in lipid profiles.[17] The evidence from clinical trials suggests that these risks are identifiable and manageable through the implementation of the mandated laboratory and clinical monitoring protocols outlined in the prescribing information. Within the context of FA, a disease that inexorably leads to severe disability, loss of independence, and premature mortality, the overall risk-benefit profile of Omaveloxolone is considered favorable for the approved patient population.[17]
While the approval of Omaveloxolone is a monumental achievement, it also marks the beginning of a new chapter in FA research and care. Several key areas require further investigation.
Published at: August 23, 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.