C25H31NO6
14484-47-0
Duchenne Muscular Dystrophy (DMD)
Deflazacort is a synthetic oxazoline derivative of prednisolone, classified as a corticosteroid prodrug. Following oral administration, it is rapidly converted to its active metabolite, 21-desacetyldeflazacort, which functions as a potent glucocorticoid receptor agonist, exerting broad anti-inflammatory and immunosuppressive effects.[1] In the United States, its sole approved indication is the treatment of Duchenne Muscular Dystrophy (DMD) in patients aged two years and older, a rare, progressive, and fatal genetic disorder characterized by muscle degeneration.[1] Clinical evidence has firmly established its efficacy in slowing the decline of muscle strength and function, delaying the loss of ambulation, preserving cardiorespiratory capacity, and reducing the incidence of scoliosis in this patient population.[1]
Despite its clinical benefits, the therapeutic profile of deflazacort is defined by a complex series of trade-offs, particularly when compared to prednisone, another corticosteroid widely used off-label for DMD. Deflazacort is associated with a more favorable profile regarding weight gain and behavioral disturbances. Conversely, it may present a higher risk of adverse effects on bone mineral density, growth velocity in children, and cataract formation.[3] This nuanced safety profile necessitates careful, individualized clinical decision-making.
The drug's regulatory and commercial history in the United States is particularly noteworthy. Although patented in 1969 and used for decades in other countries for various inflammatory conditions, it was navigated through the U.S. Food and Drug Administration (FDA) orphan drug pathway, receiving approval for DMD in 2017.[6] The subsequent announcement of a high list price generated significant controversy, positioning deflazacort as a prominent case study in the debate surrounding orphan drug incentives, drug repositioning, and pharmaceutical pricing ethics. The recent introduction of generic versions in the U.S. is expected to significantly alter its accessibility and place in therapy.[2]
Precise identification of a pharmaceutical agent is fundamental for research, clinical practice, and regulatory affairs. Deflazacort is a well-characterized small molecule with numerous identifiers across global databases and markets.
Deflazacort is known by a variety of chemical names, synonyms, and brand names worldwide. Its systematic IUPAC name isicosa-6,14,17-trien-8-yl]-2-oxoethyl] acetate.[9] A common chemical synonym is 11β,21-dihydroxy-2'-methyl-5'βH-pregna-1,4-dieno(17,16-d)oxazole-3,20-dione 21-acetate.[1]
In the United States, it is marketed under the brand names Emflaza, Jaythari, and Pyquvi.[1] Internationally, it has been available for many years under various trade names, including Calcort in the United Kingdom; Cortax, Decortil, Defcort, and Deflanil in Brazil; Moaid, Zenflav, and DefZot in India; Xalcort in Bangladesh; Zamen in Panama and Spain; and Flezacor in Honduras.[6] Table 1 provides a consolidated list of its key identifiers.
Table 1: Drug Identification and Key Database Identifiers
Identifier Type | Identifier Value | Source/Database |
---|---|---|
DrugBank ID | DB11921 | DrugBank 9 |
CAS Number | 14484-47-0 | CAS Common Chemistry 9 |
UNII | KR5YZ6AE4B | FDA Global Substance Registration System (GSRS) 9 |
Chemical Formula | C${25}H{31}NO_{6}$ | PubChem 9 |
ATC Code | H02AB13 | Drugs.com 10 |
EC Number | 238-483-7 | European Chemicals Agency (ECHA) 9 |
ChEBI ID | CHEBI:135720 | Chemical Entities of Biological Interest 9 |
ChEMBL ID | CHEMBL1201891 | ChEMBL 9 |
KEGG ID | D03671 | Kyoto Encyclopedia of Genes and Genomes 9 |
RxCUI | 22396 | NLM RxNorm 9 |
U.S. Brand Name | Emflaza | PTC Therapeutics, Inc. 11 |
U.K. Brand Name | Calcort | Wikipedia 6 |
Deflazacort is a synthetic glucocorticoid belonging to the acetonide or O-isopropylidene derivative class.[6] Its chemical formula is
C25H31NO6.[9]
Key computed physicochemical properties influence its behavior in biological systems:
These properties, particularly its polarity and hydrogen bonding capacity, are determinants of its absorption, distribution, and ability to interact with its molecular target.
The pharmacological activity of deflazacort is characteristic of the corticosteroid class, defined by its conversion to an active metabolite that modulates gene expression through the glucocorticoid receptor.
Deflazacort itself is an inactive prodrug. Following oral administration, it undergoes rapid and extensive first-pass metabolism by plasma esterases, which hydrolyze the 21-acetate group to form the pharmacologically active metabolite, 21-desacetyldeflazacort (also known as 21-desDFZ).[1] This active metabolite is responsible for the drug's systemic effects.
The mechanism of action of 21-desDFZ is as a Corticosteroid Hormone Receptor Agonist.[9] It binds with high affinity to the cytosolic glucocorticoid receptor (GR). This drug-receptor complex then translocates to the nucleus, where it functions as a ligand-dependent transcription factor. The complex can directly bind to specific DNA sequences known as glucocorticoid response elements (GREs) in the promoter regions of target genes, leading to either the transactivation (upregulation) of anti-inflammatory genes or the transrepression (downregulation) of pro-inflammatory genes.[1] This modulation of gene expression results in broad anti-inflammatory and immunosuppressive effects, which form the basis of its therapeutic utility.[2]
While these general mechanisms are well-established for all glucocorticoids, the precise molecular pathway by which deflazacort exerts its specific therapeutic effects in patients with DMD remains unknown.[1] It is hypothesized that by reducing inflammation and suppressing the immune system's activity within dystrophic muscle, deflazacort mitigates the chronic inflammatory cascade that contributes to muscle fiber necrosis and fibrosis, thereby slowing the progressive muscle damage characteristic of the disease.[9]
The pharmacokinetic profile of deflazacort describes its absorption, distribution, metabolism, and elimination (ADME), which collectively determine the onset, intensity, and duration of its therapeutic action.
Absorption: Deflazacort is rapidly absorbed from the gastrointestinal tract following oral administration. Peak plasma concentrations (Cmax) of the active metabolite, 21-desDFZ, are typically reached within 1 to 2 hours (Tmax).[1] The bioavailability of the oral tablet and oral suspension formulations is similar. Co-administration with food does not significantly alter the overall systemic absorption (as measured by the area under the curve, AUC), although a high-fat meal may reduce the peak concentration by approximately 30% and delay the time to peak by one hour.[1] For ease of administration, particularly in pediatric patients, the tablets may be crushed and mixed with applesauce without affecting absorption or bioavailability.[3]
Distribution: The active metabolite, 21-desDFZ, is approximately 40% bound to plasma proteins.[1] This is a relatively low degree of protein binding compared to some other corticosteroids, such as prednisolone, which exhibits concentration-dependent binding.[16] The volume of distribution has been determined to be 204 ± 84 L.[1]
Metabolism: As a prodrug, deflazacort's activation is the first metabolic step. The resulting active metabolite, 21-desDFZ, is subsequently metabolized in the liver, primarily by the cytochrome P450 3A4 (CYP3A4) isoenzyme, into several other inactive metabolites, including 6β-hydroxy-21-desacetyl deflazacort.[15] This heavy reliance on the CYP3A4 pathway is the mechanistic basis for deflazacort's most clinically significant drug-drug and drug-food interactions. Any substance that inhibits or induces CYP3A4 activity can profoundly alter the plasma concentrations of 21-desDFZ, necessitating dose adjustments or avoidance of co-administration.
Excretion: The elimination of deflazacort and its metabolites is predominantly via the kidneys, with approximately 68-70% of an administered dose excreted in the urine.[6] The remaining 30% is eliminated through fecal excretion.[6] The elimination process is rapid, with nearly all of the drug cleared within 24 hours post-dose.[15] The elimination half-life (
t1/2) of the active metabolite is correspondingly short, ranging from 1.1 to 1.9 hours.[6] Pharmacokinetics are not significantly altered by mild to moderate hepatic impairment or by any degree of renal impairment.[15]
Table 2: Summary of Pharmacokinetic Parameters
Parameter | Value / Description | Source(s) |
---|---|---|
Absorption | ||
Time to Peak (Tmax) | 1–2 hours | 1 |
Bioavailability | Similar for oral tablet and suspension formulations | 1 |
Food Effect | No significant effect on overall absorption (AUC); high-fat meal may reduce Cmax by ~30% | 1 |
Distribution | ||
Protein Binding | ~40% (for active metabolite 21-desDFZ) | 1 |
Metabolism | ||
Activation Pathway | Prodrug converted to active 21-desDFZ via plasma esterases | 1 |
Primary Metabolic Enzyme | CYP3A4 (for active metabolite 21-desDFZ) | 15 |
Excretion | ||
Primary Route | ~70% Renal (urinary) | 6 |
Secondary Route | ~30% Fecal | 6 |
Elimination Half-Life (t1/2) | 1.1–1.9 hours (for active metabolite 21-desDFZ) | 6 |
While deflazacort possesses broad anti-inflammatory properties applicable to many conditions, its modern clinical identity, particularly in the United States, is defined by its indication for Duchenne muscular dystrophy.
DMD is a devastating X-linked genetic disorder caused by mutations in the gene encoding the dystrophin protein. The absence of functional dystrophin destabilizes the muscle fiber membrane, leading to progressive muscle fiber necrosis, chronic inflammation, fibrosis, and eventual replacement of muscle with fat and connective tissue.[9] This pathology manifests as relentless muscle weakness, loss of ambulation in early adolescence, and ultimately, fatal cardiorespiratory failure.[9] The therapeutic rationale for using corticosteroids is to interrupt the cycle of inflammation that exacerbates muscle damage.[3]
The U.S. FDA has approved deflazacort for the treatment of Duchenne muscular dystrophy in patients 2 years of age and older, regardless of the specific genetic mutation.[2] Extensive clinical data support its efficacy in modifying the course of the disease. Key findings from clinical studies include:
Deflazacort's approval for DMD in the U.S. in 2017 represents a relatively recent chapter in its long history. The drug was first patented in 1969 and received its initial approvals for medical use in 1985.[6] For decades prior to its U.S. launch, it was widely used in Europe and other parts of the world as a first-line or alternative corticosteroid for a range of inflammatory and autoimmune diseases. These historical and ongoing off-label uses include:
Before it was formally approved by the FDA, many patients with DMD in the U.S. obtained deflazacort from international sources under personal importation policies, reflecting its long-standing recognition as a standard of care by the global neuromuscular community.[7]
The FDA's designation of deflazacort as a "first-in-class" medication for DMD is a regulatory classification that merits careful interpretation.[6] This terminology does not imply that deflazacort is a novel chemical entity or that it possesses a unique mechanism of action; it is a member of the well-established corticosteroid class. Instead, the designation signifies that it was the first drug of any kind to successfully complete the formal regulatory process and receive specific FDA approval for the DMD indication. This distinction is crucial, as it highlights how the U.S. regulatory framework, particularly the orphan drug pathway, can re-contextualize a long-established, multi-indication compound as "new" and "innovative" for a specific, underserved patient population. This re-contextualization was a key factor that enabled the market exclusivity and pricing strategy that characterized its U.S. launch.
The story of deflazacort in the United States is a complex interplay of regulatory strategy, pharmaceutical economics, and patient advocacy, serving as a significant case study in modern drug development.
The journey of deflazacort to the U.S. market was strategically managed to leverage regulatory incentives designed for rare diseases.
In stark contrast to its highly regulated and recent entry into the U.S. market, deflazacort has been an established part of the therapeutic armamentarium in Europe and other regions for decades. It has been prescribed off-label for DMD for many years, based on clinical experience and academic research.[7] The provided documentation does not indicate that deflazacort holds a formal, centralized marketing authorization from the European Medicines Agency (EMA) specifically for the DMD indication. The recent EMA recommendation for approval of vamorolone as the "first ever treatment for all patients with Duchenne" further supports the conclusion that deflazacort does not hold this specific, pan-European approval for DMD.[29] In many countries, including Canada and the United Kingdom, it has long been available as an inexpensive generic medication.[6]
The U.S. launch of deflazacort became a flashpoint in the national debate on drug pricing. The situation exemplifies the "orphan drug paradox," where incentives designed to foster innovation for rare diseases are applied to pre-existing, low-cost drugs. Marathon Pharmaceuticals executed a strategy of acquiring the rights to deflazacort, a compound with decades of real-world safety and efficacy data, and navigating it through the U.S. orphan drug pathway. This process minimized research and development risk while maximizing the commercial reward of market exclusivity.[7]
Upon receiving FDA approval, Marathon announced a list price of $89,000 per year for Emflaza. This represented an approximately 60-fold increase over the cost for patients who had been importing the drug from the U.K. or Canada, where it was available for around $1 per tablet.[6] The announcement triggered a swift and severe backlash from patient advocacy groups, physicians, and U.S. legislators, who accused the company of "price-gouging" and unethically exploiting a vulnerable patient population and the regulatory system.[6]
The intense public and political pressure led Marathon to "pause" its launch. Shortly thereafter, in March-April 2017, the company sold all rights to Emflaza to PTC Therapeutics for $140 million plus sales-based milestones.[6] PTC Therapeutics then proceeded with the commercial launch.
This episode created a significant tension between two competing public interests: enabling U.S. patients to access an effective, FDA-vetted therapy through standard prescription channels, and protecting the healthcare system and patients from what was widely perceived as predatory pricing behavior.[7] The market exclusivity granted by the orphan drug status expired for the initial indication on February 9, 2024.[27] This opened the door for generic competition, which materialized quickly. In early 2024, Aurobindo Pharma received approval for generic deflazacort tablets, and in June 2024, Cranbury Pharmaceuticals (a subsidiary of Tris Pharma) gained approval for the first generic oral suspension.[2] The entry of these lower-cost generics is expected to dramatically increase the accessibility of deflazacort for DMD patients in the U.S. and fundamentally alter the economic landscape of its use.
Proper dosing and administration are critical to maximizing the efficacy and minimizing the risks of deflazacort therapy.
Deflazacort is commercially available in the U.S. under the brand name Emflaza in two oral formulations to accommodate patient needs, particularly in the pediatric population [18]:
The dosing of deflazacort for DMD is based on patient body weight.
To ensure proper delivery and absorption, specific administration instructions should be followed.
As with all systemic corticosteroids administered for more than a few days, deflazacort must not be stopped abruptly. Doing so can precipitate acute adrenal insufficiency, a potentially fatal condition resulting from the suppression of the hypothalamic-pituitary-adrenal (HPA) axis. To prevent this, the dosage must be decreased gradually under medical supervision. Abrupt withdrawal can also trigger a steroid withdrawal syndrome, characterized by symptoms such as anorexia, nausea, fever, myalgia, and lethargy.[30]
The use of deflazacort, like all corticosteroids, is associated with a wide range of potential adverse effects and requires careful patient monitoring and risk management.
The only absolute contraindication listed for deflazacort is a known hypersensitivity to the drug substance or any of the inactive ingredients in the formulation. Rare instances of anaphylaxis have occurred in patients receiving corticosteroid therapy.[40]
The long-term use of deflazacort carries significant risks that affect multiple organ systems. These are class-wide effects for corticosteroids.
The most common adverse reactions are consistent with the known side effect profile of glucocorticoids. Data from the pivotal 12-week, placebo-controlled study in DMD provides a quantitative assessment of these risks.
Table 3: Common Adverse Reactions in DMD Clinical Trials (Deflazacort 0.9 mg/kg/d vs. Placebo at 12 Weeks) [15]
Adverse Reaction | Deflazacort 0.9 mg/kg/d (N=51) % | Placebo (N=50) % |
---|---|---|
Cushingoid appearance | 33 | 12 |
Weight increased | 20 | 6 |
Increased appetite | 14 | 2 |
Upper respiratory tract infection | 12 | 10 |
Cough | 12 | 6 |
Pollakiuria (Frequent daytime urination) | 12 | 2 |
Nasopharyngitis | 10 | 6 |
Hirsutism (Unwanted hair growth) | 10 | 2 |
Central obesity | 10 | 4 |
Erythema | 8 | 6 |
Irritability | 8 | 4 |
Rhinorrhea | 8 | 0 |
Abdominal discomfort | 6 | 2 |
The clinical use of deflazacort must account for its significant potential for interactions, which are primarily driven by its metabolism via the CYP3A4 enzyme system.
In the management of DMD, both deflazacort and prednisone/prednisolone are considered standards of care, making a direct comparison essential for clinical decision-making.[26] The choice between them is not straightforward and involves balancing nuanced differences in efficacy, potency, and, most importantly, their distinct side effect profiles.
Deflazacort is an oxazoline derivative of prednisolone, a structural modification that confers some of its unique pharmacological properties.[12] While both are potent anti-inflammatory agents, they are not equipotent on a milligram-for-milligram basis. The generally accepted dosing equivalence is that
6 mg of deflazacort has approximately the same anti-inflammatory potency as 5 mg of prednisone or prednisolone, a ratio of 1.2:1.[6] This difference in potency is reflected in the recommended daily dosages for DMD: approximately 0.9 mg/kg for deflazacort versus 0.75 mg/kg for prednisone.[32]
A key pharmacological differentiator is their effect on mineralocorticoid receptors. Deflazacort and its active metabolite have little to no affinity for these receptors, which are responsible for sodium and water retention. In contrast, prednisone possesses significant mineralocorticoid activity. This difference likely explains deflazacort's lower propensity to cause fluid retention and hypertension.[16]
Both deflazacort and prednisone have been proven to be superior to placebo in improving muscle strength and slowing disease progression in DMD. When compared directly against each other, the evidence suggests potential advantages for deflazacort in long-term functional outcomes.
The most significant differences between the two drugs lie in their side effect profiles. The clinical choice often hinges on which set of potential adverse effects is considered more manageable or less detrimental for an individual patient.
The decision between deflazacort and prednisone is therefore not a matter of selecting a superior drug, but rather a process of individualized risk-benefit assessment. It represents a clinical trade-off: one must weigh the benefits of reduced weight gain and fewer behavioral issues with deflazacort against its higher risk of cataracts and potentially greater negative impact on bone and growth. This decision must be made in consultation with the patient and their family, taking into account their specific clinical status, risk factors, and tolerance for different side effect profiles. Historically, the substantially higher cost of brand-name deflazacort in the U.S. was a significant non-clinical barrier, although the recent availability of generics may mitigate this factor.
Table 4: Comparative Profile of Deflazacort vs. Prednisone in DMD
Feature | Deflazacort | Prednisone / Prednisolone |
---|---|---|
Dosing Equivalence | ~6 mg deflazacort ≈ 5 mg prednisone (1.2:1 ratio) | Standard comparator |
Recommended DMD Dose | ~0.9 mg/kg/day | ~0.75 mg/kg/day |
Efficacy (Muscle Strength) | Effective; may offer greater benefit in preserving strength over the long term (>12 weeks) | Effective; standard of care |
Efficacy (Ambulation) | May be superior in delaying loss of ambulation and slowing decline in timed function tests | Effective at delaying loss of ambulation vs. no treatment |
Side Effect: Weight Gain | Significantly less pronounced | More pronounced; a common and significant issue |
Side Effect: Behavioral Issues | Fewer issues reported (e.g., irritability, aggression) | More issues reported |
Side Effect: Bone Health/Growth | Associated with greater growth suppression and potentially higher fracture risk in DMD cohorts | May have a less detrimental impact on bone and growth |
Side Effect: Cataracts | Higher risk of asymptomatic cataract formation | Lower risk |
Mineralocorticoid Activity | Minimal to none | Present; can contribute to fluid retention |
U.S. Cost (Historical) | Very high (brand name) | Low (generic) |
U.S. Cost (Current) | High (brand name); lower-cost generics now available | Low (generic) |
Deflazacort is a well-established corticosteroid that has been repositioned as a primary, FDA-approved therapy for Duchenne muscular dystrophy. Its efficacy in slowing the relentless progression of this disease is well-documented, offering tangible benefits in preserving muscle strength, prolonging ambulation, and protecting cardiorespiratory function. Its primary clinical advantage over the traditional standard of care, prednisone, lies in its demonstrably better profile concerning weight gain and behavioral side effects—two issues that significantly impact the quality of life for patients and their families.
However, these benefits are counterbalanced by a distinct set of risks, including a higher incidence of cataracts and potentially more severe effects on linear growth and bone health. This complex safety profile underscores that there is no universally superior corticosteroid for all patients with DMD. The therapeutic choice must be a highly individualized one, carefully weighing the specific risks and benefits for each patient.
The commercial history of deflazacort in the United States serves as a powerful and cautionary tale in pharmaceutical economics, highlighting the tensions between the laudable goals of the Orphan Drug Act and the potential for market strategies that lead to exorbitant pricing for established medicines. The recent introduction of generic alternatives marks a pivotal shift, promising to improve access and potentially rebalance the clinical decision-making process by reducing the influence of cost.
Looking forward, the role of deflazacort will continue to be defined within an evolving therapeutic landscape for DMD. The advent of novel therapies, including next-generation steroids like vamorolone, which are designed to uncouple anti-inflammatory efficacy from classic steroid side effects, as well as an expanding array of mutation-specific genetic therapies, will likely lead to more complex, combination-based treatment paradigms.[17] Future research will be essential to determine the optimal use of deflazacort—whether as a foundational monotherapy for certain patient subgroups or as a component of a multi-drug regimen—in the ongoing effort to transform the future for all individuals living with Duchenne muscular dystrophy.
Published at: September 9, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.