MedPath

Eliglustat Advanced Drug Monograph

Published:Sep 10, 2025

Generic Name

Eliglustat

Brand Names

Cerdelga

Drug Type

Small Molecule

Chemical Formula

C23H36N2O4

CAS Number

491833-29-5

Associated Conditions

Gaucher Disease, Type 1

Eliglustat (DB09039): A Comprehensive Pharmacological and Clinical Monograph

Section 1: Drug Profile and Chemical Characteristics

Eliglustat is a small molecule therapeutic agent developed as a substrate reduction therapy for a rare genetic disorder. Its pharmacological activity, clinical utility, and safety profile are intrinsically linked to its specific chemical structure and physicochemical properties. This section provides a foundational overview of its nomenclature, chemical identity, and structural features that define its function as a targeted enzyme inhibitor.

1.1 Nomenclature and Key Identifiers

For unambiguous identification across scientific, clinical, and regulatory domains, Eliglustat is cataloged under a variety of names and unique identifiers.

  • Generic Name: The officially recognized non-proprietary name for the active substance is Eliglustat.[1]
  • Brand Name: It is marketed globally under the trade name Cerdelga.[1]
  • Drug Type: Eliglustat is classified as a Small Molecule drug [User Query].
  • Developmental and Research Synonyms: During its development by Genzyme, it was referred to by codes such as GENZ-112638 and Genz-99067.[7]
  • Systematic (IUPAC) Name: The precise chemical structure is defined by its IUPAC name: N-octanamide.[1]
  • Database and Registry Identifiers: To ensure global standardization, Eliglustat is assigned unique codes in major chemical and pharmacological databases. These are essential for literature searches, regulatory tracking, and bioinformatics. A summary of these identifiers is provided in Table 1.

1.2 Physicochemical Properties and Formulation

The physical and chemical characteristics of Eliglustat dictate its formulation, stability, and route of administration. As a solid substance, it appears as a white to off-white powder.[8] Its melting point is recorded in the range of 87-92°C.[9] Solubility analysis indicates that it is soluble in dimethyl sulfoxide (DMSO) and slightly soluble in chloroform and methanol.[8] For research and storage purposes, the powder form is stable for years when stored at low temperatures, such as 2-8°C or -20°C.[7]

In clinical practice, Eliglustat is not administered as a free base but is formulated as a salt to improve its pharmaceutical properties, most commonly as Eliglustat tartrate (CAS Number: 928659-70-5) or hemitartrate.[1] The commercial product, Cerdelga, is an oral hard capsule available in 21 mg and 84 mg strengths. These capsules contain lactose monohydrate as a key excipient, a factor that is relevant for patients with rare hereditary problems of galactose intolerance.[12] The development of an effective and stable oral formulation represents a significant therapeutic advance over previous treatments that required intravenous administration.

1.3 Structural Elucidation and Stereochemistry

The therapeutic effect of Eliglustat is a direct result of its specific three-dimensional chemical structure. It is chemically classified as a carboxamide, obtained through the formal condensation of the carboxy group of octanoic acid with the primary amino group of a complex propanolamine derivative.[9] The molecule incorporates several key functional groups that define its chemical class and biological activity: a benzodioxine ring system, an N-alkylpyrrolidine moiety, a secondary alcohol, and a carboxamide linkage.[14]

Critically, the biological activity of Eliglustat is dependent on its specific stereochemistry. The IUPAC name specifies the (1R,2R) stereoisomers, indicating that the precise spatial arrangement of atoms at its two chiral centers is essential for effective binding to its molecular target.[1] This stereospecificity is a common feature of potent and selective drugs that interact with biological macromolecules.

The rational design of this molecule is central to its mechanism of action. Eliglustat functions as a "ceramide mimic," a structural analog of the endogenous lipid ceramide.[15] This mimicry is achieved through its distinct structural domains. The long, flexible eight-carbon alkyl chain derived from octanoic acid mimics the fatty acyl tail of ceramide. The more complex headgroup, containing the benzodioxine, hydroxyl, and pyrrolidinyl groups, mimics the sphingoid base of ceramide. This structural homology allows Eliglustat to fit into the ceramide-binding site of its target enzyme, glucosylceramide synthase (GCS), and act as a potent and competitive inhibitor.[16] This deliberate structure-activity relationship is the chemical foundation of its therapeutic efficacy in reducing the production of glucosylceramide.

Table 1: Key Identifiers and Physicochemical Properties of Eliglustat

PropertyValueSource(s)
DrugBank IDDB090391
CAS Number (Free Base)491833-29-51
PubChem CID236527311
UNIIDR40J4WA671
Chemical FormulaC23​H36​N2​O4​1
Molecular Weight404.54 g/mol1
InChIKeyFJZZPCZKBUKGGU-AUSIDOKSSA-N1
AppearanceWhite to off-white solid powder8
ATC CodeA16AX101

Section 2: Developmental and Regulatory Trajectory

The journey of Eliglustat from an academic concept to a globally approved medication is a case study in modern drug development, marked by scientific innovation, strategic corporate decisions, and a paradigm-shifting regulatory approach that has embraced personalized medicine.

2.1 Discovery, Preclinical Development, and Manufacturer

The conceptual origins of Eliglustat trace back to 1982, when Dr. Norman Radin at the University of Michigan began investigating the possibility of treating lysosomal storage diseases by inhibiting the synthesis of the accumulating lipid substrates.[1] This represented a novel therapeutic strategy, later termed Substrate Reduction Therapy (SRT). In the mid-1990s, this research, in collaboration with the laboratory of Dr. Jim Shayman, yielded several promising candidate inhibitors of glucosylceramide synthase.[1]

Genzyme Corporation, a leader in therapies for rare diseases, initially passed on these candidates. However, the company licensed the patents from the University of Michigan in 2000, reportedly spurred by news of a competitor's progress in developing a new treatment for Gaucher's disease.[1] The subsequent development process was lengthy, with a 14-year gap between the licensing agreement and final FDA approval. Dr. Shayman speculated that this delay may have been influenced by a degree of corporate reluctance within Genzyme to develop a new oral drug that would directly compete with its own highly successful flagship product, the intravenous enzyme replacement therapy (ERT) imiglucerase (brand name Cerezyme).[1] Today, the drug is marketed by Sanofi, which acquired Genzyme.[19]

2.2 Global Regulatory Approvals and Orphan Drug Designation

Eliglustat has received marketing authorization from major regulatory agencies worldwide, solidifying its role as a key treatment for Gaucher disease type 1 (GD1).

  • United States Food and Drug Administration (FDA): Genzyme submitted the New Drug Application (NDA) for Eliglustat on September 19, 2013.[6] Following a priority review, the FDA approved Cerdelga on August 19, 2014.[1] The approved indication is for the long-term treatment of adult patients with GD1 who are determined to be CYP2D6 extensive metabolizers (EMs), intermediate metabolizers (IMs), or poor metabolizers (PMs) through the use of an FDA-cleared test.[20]
  • European Medicines Agency (EMA): In Europe, the Committee for Medicinal Products for Human Use (CHMP) issued a positive opinion on November 20, 2014, leading to a full marketing authorization valid throughout the European Union on January 19, 2015.[19] The EMA's indication is broader than the FDA's, including not only adults but also pediatric patients from 6 years of age who weigh at least 15 kg and are stable on ERT.[13]
  • Orphan Drug Status: Recognizing the rarity of Gaucher disease, both agencies granted Eliglustat orphan drug designation, which provides incentives for the development of treatments for rare conditions. The EMA designated it as an orphan medicine on December 4, 2007, and the FDA followed on September 17, 2008.[19] This status provided market exclusivity for a period following approval.[20]
  • Legal Status: Consistent with its potent pharmacological effects and complex prescribing requirements, Eliglustat is classified as a prescription-only medication in all major markets, including the United States (℞-only), the European Union (Rx-only), the United Kingdom (POM), Canada (℞-only), and Australia (S4).[1]

The regulatory approval of Eliglustat is particularly noteworthy for its explicit codification of a personalized medicine approach. The labels from both the FDA and EMA do not merely recommend or suggest genetic testing; they mandate it as a required patient selection step prior to initiating therapy.[13] This decision was based on extensive clinical data demonstrating that the drug's pharmacokinetic profile, efficacy, and particularly its safety, are inextricably linked to a patient's genetically determined CYP2D6 metabolizer status.[17] By making an

a priori pharmacogenomic test a formal gateway to treatment, regulatory bodies have elevated this practice from a supplementary consideration to an indispensable component of the drug's approved use. This sets a significant precedent for future therapeutics with similarly pronounced pharmacogenomic dependencies, signaling a regulatory shift towards embedding personalized medicine directly into prescribing protocols.

Section 3: Pharmacodynamics: Mechanism of Substrate Reduction

The therapeutic efficacy of Eliglustat in Gaucher disease is derived from its precise and potent modulation of a key metabolic pathway. As a Substrate Reduction Therapy (SRT), it addresses the underlying pathophysiology of the disease at its source by limiting the production of the accumulating toxic substrate. Recent research has also begun to uncover additional cellular mechanisms that may contribute to its biological effects.

3.1 Primary Mechanism: Inhibition of Glucosylceramide Synthase (GCS)

Eliglustat's primary mechanism of action is the potent and selective inhibition of the enzyme glucosylceramide synthase (GCS), also known as ceramide glucosyltransferase (EC 2.4.1.80).[1] GCS is a pivotal enzyme located in the Golgi apparatus that catalyzes the first committed step in the biosynthesis of the vast majority of glycosphingolipids (GSLs).[16] Specifically, it facilitates the transfer of a glucose molecule from an activated donor, UDP-glucose, to the lipid ceramide, thereby forming glucosylceramide (GlcCer).[16]

By inhibiting GCS, Eliglustat effectively reduces the overall rate of GlcCer synthesis within the cell.[17] This "upstream" intervention decreases the quantity of GlcCer that is subsequently trafficked to the lysosomes for catabolism, thus lessening the metabolic burden on the cell's degradative machinery.[7]

3.2 Molecular Specificity and In Vitro Potency

The clinical utility of Eliglustat is underpinned by its high potency and specificity for its target enzyme. In vitro assays have consistently demonstrated its potent inhibitory activity, with reported half-maximal inhibitory concentration (IC50​) values of 10 ng/mL, 20 nM in intact Madin-Darby Canine Kidney (MDCK) cells, and 24 nM in other cell-based assays.[7] This high potency ensures that effective enzyme inhibition can be achieved at clinically relevant plasma concentrations.

Furthermore, this inhibition is highly specific. Studies have shown that Eliglustat has minimal to no off-target activity against a panel of other related enzymes, including other glycosidases like α-glucosidase I and II, the lysosomal and non-lysosomal glucosylceramidases that degrade GlcCer, and digestive enzymes such as sucrase and maltase.[7] This high degree of specificity is crucial for minimizing the potential for off-target adverse effects, distinguishing it from less specific inhibitors.

3.3 Pathophysiological Impact in Gaucher Disease

Gaucher disease is a classic lysosomal storage disorder resulting from inherited mutations in the GBA1 gene, which leads to a deficiency of the lysosomal enzyme acid β-glucosidase (also known as glucocerebrosidase).[14] The function of this enzyme is to break down GlcCer into glucose and ceramide. In its absence, GlcCer accumulates progressively within the lysosomes, particularly in cells of the monocyte-macrophage lineage.[16] These engorged, lipid-laden macrophages, termed "Gaucher cells," are the pathological hallmark of the disease. They infiltrate various organs, primarily the spleen, liver, and bone marrow, leading to the characteristic clinical manifestations of hepatosplenomegaly, anemia, thrombocytopenia, and skeletal disease.[16]

Eliglustat's mechanism as an SRT directly counteracts this pathophysiology. By reducing the synthesis of GlcCer, it lowers the substrate load on the deficient lysosomal enzyme system.[24] This reduction in substrate influx prevents the massive accumulation of GlcCer, leading to a decrease in the number and size of Gaucher cells in affected tissues and a subsequent alleviation of the downstream organ damage and hematological abnormalities.[8] This approach is fundamentally different from ERT, which works by supplying a recombinant version of the missing enzyme to clear the already-accumulated substrate. SRT, by contrast, aims to restore metabolic balance by preventing the accumulation in the first place.

3.4 Emerging Research on Novel Cellular Mechanisms

While the primary mechanism of GCS inhibition is well-established, emerging preclinical evidence suggests that the biological activities of Eliglustat may be more complex. A recent study investigating its effects in preclinical models of multiple myeloma (MM), a cancer of plasma cells that often causes severe bone disease, identified a novel mechanism of action.[27] In this context, Eliglustat was found to act as an inhibitor of autophagy flux specifically in bone-resorbing cells known as osteoclasts.

Autophagy is a fundamental cellular process for degrading and recycling cellular components. By inhibiting this process in osteoclasts, Eliglustat was shown to alter the lipid composition and fluidity of the plasma membrane, which in turn reduced osteoclast formation and function. The ultimate effect in these models was an increase in bone mass and a reduction in myeloma-associated bone disease.[27]

This discovery carries significant implications. Firstly, it suggests that the full mechanistic profile of Eliglustat may not be limited to GCS inhibition and that some of its therapeutic effects or even side effects, potentially in bone, could be mediated through this autophagy pathway. Secondly, it opens the door to potential therapeutic repurposing. A drug designed for a rare genetic lipid storage disorder might find new applications in treating more common conditions characterized by excessive bone loss or pathological autophagy, such as certain cancers or metabolic bone diseases. This highlights the value of continued investigation into the broader cellular impacts of targeted therapies.

Section 4: Pharmacokinetics: A Genotype-Dependent Profile

The absorption, distribution, metabolism, and excretion (ADME) profile of Eliglustat is a defining feature of the drug, with profound clinical implications. Its pharmacokinetics are characterized by a critical dependence on the function of specific cytochrome P450 enzymes, which are subject to significant genetic variation in the population. This relationship between genetics and drug exposure is the central determinant of Eliglustat's efficacy and safety.

4.1 Absorption, Distribution, and Administration

Eliglustat is administered orally in the form of a hard capsule.[1] The prescribing information provides specific instructions for its administration to ensure optimal absorption and avoid interactions. The capsules must be swallowed whole and should not be crushed, opened, or dissolved.[13] This suggests that the formulation is designed for controlled release or to protect the active ingredient from the gastric environment. The drug can be taken with or without food, indicating that food does not significantly impact its overall bioavailability.[13]

A crucial administration instruction is the strict avoidance of grapefruit and grapefruit juice.[13] This is a classic food-drug interaction warning related to the potent inhibition of the CYP3A4 enzyme by compounds in grapefruit, which would otherwise lead to dangerously elevated plasma concentrations of Eliglustat. Preclinical studies evaluating its distribution and pharmacokinetic properties in various animal species (mouse, rat, dog, and monkey) found that its ADME characteristics were generally consistent across species, providing a reliable basis for toxicological assessment.[29]

4.2 Metabolism: The Central Roles of Cytochrome P450 Enzymes CYP2D6 and CYP3A4

Following absorption, Eliglustat undergoes extensive hepatic metabolism, which is the primary route of its clearance from the body.[14] This metabolism is almost entirely dependent on the cytochrome P450 (CYP) superfamily of enzymes.

  • Primary Pathway (CYP2D6): The vast majority of Eliglustat metabolism is mediated by the CYP2D6 enzyme.[14] Quantitative analysis estimates that approximately 86% of the drug's clearance is attributable to this pathway (fraction metabolized, fm​≈0.86).[22]
  • Secondary Pathway (CYP3A4): A smaller, but still significant, portion of metabolism is handled by the CYP3A4 enzyme, accounting for the remaining clearance (fm​≈0.14).[15]

The metabolites produced through these pathways are known to be inactive, meaning the parent drug is the sole active entity and its clearance represents a true detoxification process.[15] This heavy reliance on a single primary metabolic enzyme, CYP2D6, makes the drug's pharmacokinetic profile exceptionally sensitive to any factors that alter the function of this enzyme.

4.3 The Clinical Impact of CYP2D6 Genetic Polymorphisms

The gene encoding the CYP2D6 enzyme is highly polymorphic in the human population, leading to wide inter-individual variability in enzyme activity. This genetic variation is the single most important factor determining a patient's exposure to Eliglustat. Based on their CYP2D6 genotype, individuals are classified into distinct phenotypes [15]:

  • Poor Metabolizers (PMs): Carry two non-functional alleles, leading to absent or minimal enzyme activity.
  • Intermediate Metabolizers (IMs): Carry combinations of alleles that result in decreased enzyme activity.
  • Extensive Metabolizers (EMs): Have two fully functional alleles, representing the "normal" metabolic capacity.
  • Ultra-rapid Metabolizers (URMs): Carry multiple copies of functional alleles, leading to exceptionally high enzyme activity.

These genetic differences have direct and predictable clinical consequences for Eliglustat therapy. In PMs and IMs, the reduced clearance leads to significantly higher plasma concentrations and drug exposure for a given dose. Conversely, in URMs, the drug is cleared so rapidly that it is difficult to achieve and maintain concentrations within the therapeutic window.[15] Consequently, treatment with Eliglustat is not recommended for URMs due to the high likelihood of therapeutic failure. To prevent toxicity from overexposure, PMs require a reduced dose (84 mg once daily), while EMs and IMs receive the standard dose (84 mg twice daily) to achieve the target exposure.[15]

4.4 Elimination and Half-Life

After being metabolized in the liver, the inactive metabolites of Eliglustat are eliminated from the body via both the urinary and gastrointestinal (fecal) routes.[15] Preclinical studies in animals reported relatively short terminal half-lives, ranging from approximately 0.3 to 1.4 hours, suggesting rapid clearance in these species.[29] However, pharmacokinetic data from human trials have shown considerable variability between patients, which is a direct reflection of the differing metabolic capacities dictated by CYP2D6 genotype.[31]

The entire risk profile and management strategy for Eliglustat can be understood as a direct cascade of events originating from its metabolic pathway. The chain of causality begins with the drug's heavy reliance on CYP2D6 for clearance. This creates a vulnerability to the well-known genetic polymorphisms of the CYP2D6 gene, as well as to the many common medications that inhibit CYP2D6 or the secondary CYP3A4 pathway. Either a genetic deficiency in enzyme function (as seen in PMs and IMs) or competitive inhibition by a co-administered drug results in decreased clearance and, consequently, substantially elevated plasma concentrations (Cmax​ and AUC).[22] This risk of overexposure is not benign; pharmacodynamic and safety pharmacology studies have established a clear link between high plasma concentrations of Eliglustat (e.g., >250-500 ng/mL) and clinically significant prolongation of cardiac electrical intervals, specifically the PR, QRS, and QTcF intervals.[14] This electrophysiological effect creates the primary safety concern for the drug: a heightened risk of potentially life-threatening cardiac arrhythmias. This single, linear progression—from metabolism to exposure to cardiac risk—necessitates the complex, multi-layered risk mitigation strategy that defines Eliglustat's clinical use, including mandatory genotyping, genotype-based dosing, an extensive list of drug-drug interaction contraindications, and warnings for patients with underlying cardiac conditions.

Section 5: Clinical Efficacy in Gaucher Disease Type 1

The approval of Eliglustat as a treatment for Gaucher disease type 1 (GD1) was based on a robust clinical development program that demonstrated its efficacy in improving the key clinical manifestations of the disease. The pivotal trials were strategically designed to establish its utility both as a primary therapy for untreated patients and as a convenient oral alternative for patients already stabilized on standard intravenous therapy.

5.1 Evidence from Pivotal Phase 3 Clinical Trials

The core evidence for Eliglustat's efficacy comes from two large, randomized, controlled Phase 3 trials, supported by data from an earlier Phase 2 study.[33]

  • ENGAGE Trial (NCT00891202): This was a pivotal study designed to evaluate the efficacy and safety of Eliglustat in "treatment-naïve" adult patients with GD1 who had not previously received any therapy for the disease. This trial was crucial for establishing Eliglustat as a first-line treatment option.[33]
  • ENCORE Trial (NCT00943111): This study enrolled adult GD1 patients whose disease had already been stabilized through long-term treatment with enzyme replacement therapy (ERT). Patients were randomized to either switch to oral Eliglustat or continue their intravenous ERT. The primary objective was to demonstrate that Eliglustat was effective in maintaining disease stability, positioning it as a viable alternative to infusions.[33]

Additional supportive data were provided by the Phase 2 trial GZGD00304 and the Phase 3 EDGE trial (GZGD03109), which further characterized the drug's safety and efficacy profile.[33]

5.2 Analysis of Key Efficacy Endpoints

Across these clinical trials, Eliglustat demonstrated statistically significant and clinically meaningful improvements in the established markers of disease activity in GD1.[1]

  • Visceral Improvements: In the ENGAGE trial, treatment-naïve patients receiving Eliglustat for 9 months experienced a mean reduction in spleen volume of 28%, compared to a 2% increase in the placebo group. Similar significant reductions in liver volume were also observed.[1] These results confirm the drug's ability to reverse the organomegaly that is a hallmark of GD1.
  • Hematological Improvements: Patients in the ENGAGE trial also showed significant improvements in hematological parameters, including increases in hemoglobin levels (addressing anemia) and platelet counts (addressing thrombocytopenia).[1]
  • Maintenance of Disease Stability: In the ENCORE trial, which evaluated the switch from ERT, Eliglustat successfully maintained disease stability in the majority of patients. After one year, 85% of patients who switched to Eliglustat remained stable, a result that compared favorably with the 94% stability rate observed in patients who continued on their ERT infusions.[19] This demonstrated that for most stable patients, the transition to an oral therapy could be made without compromising disease control.

5.3 Therapeutic Role: First-Line Oral Therapy and Transition from ERT

Based on this strong clinical evidence, Eliglustat has been established as a key component of the GD1 treatment armamentarium. It is approved as a first-line oral therapy for eligible adult patients, offering a new initial treatment option alongside the traditional ERTs.[22]

Perhaps its most significant impact has been as a non-invasive, orally administered alternative to the standard-of-care ERTs, which include imiglucerase, velaglucerase alfa, and taliglucerase alfa.[16] These biologic therapies require lifelong, regular (typically bi-weekly) intravenous infusions, which can be burdensome for patients, impacting their quality of life, work, and travel. The availability of an effective oral agent like Eliglustat represents a major therapeutic advance, offering patients greater convenience and autonomy in managing their chronic condition.[17]

Section 6: Clinical Application and Prescribing Guidelines

The clinical application of Eliglustat is highly structured and requires a meticulous, multi-step approach to patient selection and management. The prescribing guidelines are designed to maximize therapeutic benefit while proactively mitigating the significant risks associated with its genotype-dependent metabolism and potential for drug interactions.

6.1 Patient Selection: The Mandate for CYP2D6 Genotyping

The foundational step in prescribing Eliglustat is the determination of the patient's CYP2D6 metabolizer status. This is not a recommendation but a strict requirement outlined by regulatory agencies.

  • Mandatory Testing: Before initiating therapy, all patients must undergo genotyping for the CYP2D6 gene using an FDA- or EMA-cleared test.[13] This test determines the patient's innate capacity to metabolize the drug.
  • Eligible Patient Population: Treatment with Eliglustat is indicated only for patients who are identified as Poor Metabolizers (PMs), Intermediate Metabolizers (IMs), or Extensive Metabolizers (EMs).[12]
  • Ineligible Patient Populations: The drug is not recommended for two groups. Firstly, Ultra-rapid Metabolizers (URMs) are excluded because their high enzyme activity is likely to clear the drug too quickly, preventing them from achieving adequate plasma concentrations for a therapeutic effect. Secondly, a specific dose cannot be recommended for patients whose genotype cannot be determined (termed "indeterminate metabolizers").[12]

6.2 Genotype-Specific Dosing Regimens and Administration

Once a patient's CYP2D6 phenotype is established, the dosing regimen is tailored specifically to their metabolic capacity to ensure drug exposure is maintained within the optimal therapeutic window. The adult dosing recommendations are as follows [15]:

Table 2: Dosing Recommendations Based on CYP2D6 Metabolizer Status and Concomitant Medications

CYP2D6 PhenotypeConcomitant InhibitorsHepatic/Renal StatusRecommended Eliglustat Dose / Action
Extensive (EM) or Intermediate (IM)NoneNormal84 mg twice daily
Poor (PM)NoneNormal84 mg once daily
EM or IMStrong or Moderate CYP2D6 InhibitorNormal84 mg once daily
EMStrong or Moderate CYP3A InhibitorNormal84 mg once daily
EM or IMStrong/Moderate CYP2D6 Inhibitor + Strong/Moderate CYP3A InhibitorNormalContraindicated
IM or PMStrong CYP3A InhibitorNormalContraindicated
IM or PMAny degree of hepatic or renal impairmentN/AContraindicated or Avoid
EMModerate or Severe Hepatic ImpairmentN/AContraindicated
EMESRD (CrCl <15 mL/min)N/AAvoid
Ultra-rapid (URM) or IndeterminateN/AN/ANot Recommended / Contraindicated

Note: This table is a simplified summary. Clinicians must consult the full prescribing information for complete details on all combinations of inhibitors and specific patient populations. [12]

In the European Union, where the drug is approved for pediatric use, dosing is also dependent on both genotype and body weight.[13]

6.3 Management in Special Populations: Hepatic and Renal Impairment

The safe use of Eliglustat is further complicated by the presence of organ dysfunction, as both the liver and kidneys play roles in the drug's metabolism and excretion.

  • Hepatic Impairment: The liver is the primary site of Eliglustat metabolism. Therefore, hepatic impairment can significantly reduce its clearance and increase exposure. The recommendations are highly specific and depend on an interplay between the degree of liver dysfunction (as measured by the Child-Pugh score) and the patient's CYP2D6 genotype. For instance, use is contraindicated in EMs with moderate or severe hepatic impairment and in all IMs and PMs with any degree of hepatic impairment.[12]
  • Renal Impairment: While not the primary route of elimination for the parent drug, renal function is still a consideration. Use of Eliglustat should be avoided in patients with End-Stage Renal Disease (ESRD, CrCl <15 mL/min). While no dose adjustment is required for EMs with mild to severe renal impairment, use is generally not recommended for IMs or PMs with any degree of renal impairment due to limited data and the potential for accumulation.[12]

The clinical decision-making process for prescribing Eliglustat is therefore exceptionally complex. It is not a linear or single-factor assessment. Instead, it requires the clinician to navigate a multi-dimensional matrix of interacting variables. The four key pillars of this assessment are: (1) the patient's immutable CYP2D6 genotype, which sets the baseline metabolic capacity; (2) a comprehensive review of all concomitant medications to identify CYP inhibitors or inducers; (3) an evaluation of hepatic function; and (4) an assessment of renal function. A change in any one of these variables can have a cascading effect, potentially requiring a dose adjustment or, in many cases, rendering the drug contraindicated for that patient. For example, an EM with normal organ function on the standard dose may need a dose reduction if they develop mild hepatic impairment and are prescribed a weak CYP3A inhibitor—a three-factor interaction.[12] This intricate decision algorithm underscores the drug's narrow therapeutic index and high-risk metabolic profile, demanding a high level of clinical vigilance from the prescribing physician.

Section 7: Safety, Tolerability, and Risk Management

The safety profile of Eliglustat is generally considered manageable for a chronic therapy, but it is characterized by a significant and well-defined cardiovascular risk that dictates much of its clinical management. A comprehensive risk management strategy, embedded within the drug's label, is essential for its safe use.

7.1 Profile of Common and Serious Adverse Events

In pivotal clinical trials, Eliglustat was reasonably well-tolerated. The most frequently reported adverse reactions, occurring in 10% or more of patients, were generally mild to moderate and often transient. These include fatigue, headache, nausea, diarrhea, back pain, pain in the extremities, and upper abdominal pain.[21]

Serious adverse events were less common but did occur. Syncope (fainting) was reported in a small number of patients in clinical trials, a symptom that is particularly noteworthy given the drug's potential effects on cardiac conduction.[28] With regard to organ-specific toxicity, there has been no signal for significant hepatotoxicity. In placebo-controlled trials, liver test abnormalities were no more common with Eliglustat than with placebo, and no cases of acute liver injury with jaundice have been attributed to the drug in post-marketing experience. Consequently, its likelihood score for causing clinically apparent liver injury is rated as E (unlikely, though clinical experience remains limited).[25]

7.2 Cardiovascular Risk: ECG Interval Prolongation and Arrhythmias

The primary and most serious safety concern associated with Eliglustat is its potential to cause cardiac arrhythmias at elevated plasma concentrations.[21]

  • Mechanism of Cardiac Risk: The underlying mechanism is the drug's effect on cardiac electrophysiology. At substantially increased concentrations, Eliglustat is predicted to cause dose-dependent prolongation of key intervals on an electrocardiogram (ECG), including the PR interval (atrioventricular conduction), the QRS duration (ventricular depolarization), and the QTc interval (ventricular repolarization).[14] Prolongation of these intervals, particularly the QTc interval, is a known risk factor for life-threatening ventricular arrhythmias such as Torsades de Pointes.
  • Pharmacological Basis: This risk is supported by nonclinical data. In vitro studies demonstrated that Eliglustat inhibits the hERG potassium channel, a key channel involved in cardiac repolarization, with an IC50​ of 0.35 µg/mL.[29] Furthermore, pharmacokinetic/pharmacodynamic (PK/PD) modeling predicts that at a plasma concentration of 500 ng/mL, Eliglustat would cause clinically relevant increases in PR, QRS, and QTcF intervals of 22 msec, 7 msec, and 13 msec, respectively.[14]
  • Clinical Monitoring: Due to this risk, an ECG is often recommended before initiating therapy to establish a baseline.[28] Patients must be counseled to report any symptoms suggestive of an arrhythmia, such as palpitations, dizziness, or syncope, to their healthcare provider immediately.[35]

7.3 Contraindications and Precautionary Measures

To mitigate the cardiovascular risk, the prescribing information for Eliglustat includes a comprehensive list of contraindications and warnings.

  • Patients with Pre-existing Cardiac Conditions: The use of Eliglustat has not been studied in patients with underlying heart disease. Therefore, its use should be avoided in patients with pre-existing conditions such as congestive heart failure, recent acute myocardial infarction, bradycardia, heart block, or ventricular arrhythmia. It is also not recommended for patients with congenital long QT syndrome.[12]
  • Concomitant Antiarrhythmic Drugs: To avoid additive effects on cardiac conduction, concomitant use of Eliglustat with Class IA (e.g., quinidine, procainamide) or Class III (e.g., amiodarone, sotalol) antiarrhythmic agents is not recommended.[14]

Despite the well-defined and serious nature of this cardiac risk, Eliglustat does not carry a "Black Box Warning" in its FDA-approved label.[43] This represents a significant regulatory decision. Black Box Warnings are typically reserved for drugs with the most serious or life-threatening risks. The absence of such a warning for Eliglustat suggests that regulators deemed the risk to be manageable through other means. The risk of arrhythmia is not universal but is confined to predictable and identifiable scenarios: patients with specific genetic profiles (PMs), those taking interacting medications, or those with organ impairment. The regulatory strategy appears to have been to proactively "design out" these high-risk scenarios through the label itself. By mandating pharmacogenomic testing and establishing an extensive and highly specific list of contraindications and dose adjustments, the label creates a framework where, if followed precisely, the risk is theoretically minimized for the intended, carefully selected patient population. This places a significant burden of responsibility on the prescribing physician to strictly adhere to the complex label to ensure patient safety.

Section 8: Drug-Drug Interaction Profile

Eliglustat is subject to numerous, complex, and clinically significant drug-drug interactions (DDIs). These interactions are a central feature of its pharmacological profile and are critical considerations for its safe administration. The interactions are primarily metabolic and can be bidirectional, meaning Eliglustat concentrations can be altered by other drugs, and Eliglustat itself can alter the concentrations of other drugs.

8.1 Interactions with CYP2D6 and CYP3A4 Inhibitors and Inducers

As a substrate that is extensively metabolized by CYP2D6 and to a lesser extent by CYP3A4, Eliglustat's plasma concentration is highly susceptible to modulation by other drugs that affect these enzymes.[15]

  • CYP Inhibitors: Co-administration with drugs that inhibit CYP2D6 or CYP3A4 can dramatically increase Eliglustat exposure, thereby amplifying the risk of cardiac toxicity.
  • Strong CYP2D6 inhibitors (e.g., paroxetine, fluoxetine, bupropion) can increase Eliglustat AUC by 7- to 9-fold.[13]
  • Strong CYP3A4 inhibitors (e.g., ketoconazole, clarithromycin, and compounds in grapefruit juice) can increase Eliglustat AUC by approximately 4-fold.[21]
  • The concurrent use of inhibitors of both pathways is particularly dangerous and is contraindicated in many patient populations.[12]
  • CYP Inducers: Conversely, co-administration with strong CYP3A inducers (e.g., rifampin, carbamazepine, phenytoin, St. John's wort) can significantly decrease Eliglustat plasma concentrations by accelerating its metabolism. This may lead to a loss of therapeutic efficacy and is therefore not recommended.[15]

The clinical management of these interactions is highly complex and is strictly dependent on the patient's CYP2D6 metabolizer status. The prescribing information provides detailed, genotype-specific instructions that range from dose reduction to absolute contraindication (as summarized in Table 2). The extensive list of potential interactions, with over 368 drugs identified by one database, necessitates a meticulous review of a patient's entire medication list, including over-the-counter products and herbal supplements, before and during therapy.[44]

8.2 Eliglustat as an Inhibitor of CYP2D6 and P-glycoprotein (P-gp)

The potential for DDIs is bidirectional, as Eliglustat itself acts as an inhibitor of other key drug disposition pathways. In vitro data and clinical studies have shown that Eliglustat is an inhibitor of both the CYP2D6 enzyme and the P-glycoprotein (P-gp) efflux transporter.[13]

This means that Eliglustat can increase the plasma concentrations of other medications that are substrates of these pathways, potentially leading to their toxicity.

  • CYP2D6 Substrates: Caution and potential dose reduction are advised when co-administering Eliglustat with sensitive CYP2D6 substrates, such as the beta-blocker metoprolol, certain phenothiazines, and tricyclic antidepressants.[37]
  • P-gp Substrates: Eliglustat can increase the exposure of P-gp substrates. For example, it is recommended that the dose of digoxin be reduced by 30% when initiating Eliglustat therapy. Monitoring or dose reduction may also be necessary for other P-gp substrates like dabigatran and colchicine.[37]

8.3 A Framework for Clinical Management of Significant Interactions

Given the complexity and critical nature of these interactions, clinicians must follow a structured approach. The management strategy is built upon the patient's CYP2D6 genotype and involves consulting detailed tables in the prescribing information to determine the appropriate action for any given co-administered drug. A summary of key interactions and their management is presented in Table 3.

Table 3: Summary of Clinically Significant Drug-Drug Interactions with Eliglustat

Interacting Drug/ClassMechanism of InteractionEffect on Eliglustat / Other DrugClinical Management Recommendation
Paroxetine, Bupropion, FluoxetineStrong CYP2D6 Inhibition↑ Eliglustat exposure (7-9 fold)Reduce Eliglustat dose to 84 mg once daily in EMs and IMs.
Ketoconazole, Clarithromycin, Grapefruit JuiceStrong CYP3A4 Inhibition↑ Eliglustat exposure (4-fold)Reduce Eliglustat dose to 84 mg once daily in EMs. Contraindicated in IMs and PMs.
Rifampin, Carbamazepine, St. John's WortStrong CYP3A4 Induction↓ Eliglustat exposureConcomitant use is not recommended due to risk of therapeutic failure.
Amiodarone, SotalolClass III AntiarrhythmicsAdditive QTc prolongation riskConcomitant use is not recommended.
DigoxinP-gp Substrate↑ Digoxin exposureReduce digoxin dose by 30% upon initiating Eliglustat and monitor serum levels.
MetoprololCYP2D6 Substrate↑ Metoprolol exposureMonitor for increased effects (e.g., bradycardia); consider dose reduction of metoprolol.

Note: This table highlights examples and is not exhaustive. Always refer to the full, current prescribing information for complete guidance. [13]

Section 9: Comparative and Economic Analysis

Eliglustat's entry into the therapeutic landscape for Gaucher disease provided a new treatment modality that must be evaluated in the context of existing therapies and the economic realities of the orphan drug market.

9.1 Therapeutic Comparison with Enzyme Replacement and Other Substrate Reduction Therapies

The primary comparator for Eliglustat is Enzyme Replacement Therapy (ERT), the long-standing standard of care for GD1.

  • Comparison with ERT (Imiglucerase, Velaglucerase alfa, Taliglucerase alfa): The most profound advantage of Eliglustat over ERT is its oral route of administration, which liberates patients from the lifelong burden of regular intravenous infusions.[17] This represents a substantial improvement in convenience and quality of life. In terms of efficacy, the ENCORE trial demonstrated that Eliglustat was effective at maintaining disease stability in patients who switched from ERT, with 85% of patients remaining stable after one year. While this was a strong result, it was numerically slightly lower than the 94% stability rate seen in patients who continued on ERT, suggesting that a small subset of patients may respond more robustly to ERT.[19] The choice between SRT and ERT thus involves a clinical discussion weighing the convenience of an oral pill against the extensive long-term data and potentially slightly higher stability rates of intravenous ERT.
  • Comparison with Miglustat: Miglustat is another approved oral SRT for GD1. The provided data does not contain a direct head-to-head comparison of the efficacy or safety profiles of Eliglustat and Miglustat, but Eliglustat is often considered a first-line oral option due to its specificity and tolerability profile.[16]

9.2 Economic Considerations and Market Positioning as an Orphan Drug

The pricing of Eliglustat reflects the economic dynamics of the orphan drug market, where therapies for rare diseases command high prices due to small patient populations and the high cost of development.

  • Cost: At the time of its launch in 2014, the annual cost of treatment with oral Eliglustat was reported to be approximately $310,250.[1] This price was set to be highly comparable to the existing cost of intravenous ERT with Cerezyme (imiglucerase), which was approximately $300,000 per year for a typical patient.[1]
  • Orphan Drug Pricing Strategy: This pricing strategy is notable because, as a small molecule, the manufacturing costs for Eliglustat are significantly lower than those for a complex recombinant biologic like imiglucerase.[1] The decision to price it at parity with the existing standard of care, rather than at a discount, reflects a value-based pricing model. The value proposition is the added convenience of oral administration. Genzyme (now Sanofi) has maintained that such pricing for orphan drugs, which are most often paid for by insurers, is necessary to sustain the financial viability of developing treatments for rare diseases.[1]

Section 10: Conclusion and Future Perspectives

Eliglustat represents a significant milestone in the treatment of Gaucher disease type 1 and a landmark achievement in the clinical implementation of personalized medicine. Its development and approval have provided a valuable oral therapeutic option, fundamentally altering the management landscape for this rare genetic disorder. However, its use demands a sophisticated understanding of its complex pharmacology, and ongoing research continues to explore its full therapeutic potential.

10.1 Synthesis of Eliglustat's Role in the Management of Gaucher Disease

Eliglustat has been successfully established as a first-line oral Substrate Reduction Therapy for eligible adults with GD1. Its primary contribution to clinical practice is offering a non-invasive alternative to lifelong intravenous ERT, thereby significantly enhancing patient convenience and quality of life. Clinical trials have robustly demonstrated its efficacy in improving the key visceral and hematological manifestations of the disease and in maintaining stability in patients transitioning from ERT.

However, the therapeutic benefits of Eliglustat are inextricably linked to a rigorous, genotype-driven approach to patient management. Its heavy reliance on CYP2D6 for metabolism creates a narrow therapeutic window and a significant risk of cardiac arrhythmia in the event of overexposure. Consequently, its safe and effective use is critically dependent on mandatory pharmacogenomic testing, careful genotype-specific dosing, and meticulous management of a vast and complex profile of potential drug-drug interactions. Eliglustat is, therefore, a prime exemplar of modern pharmacotherapy, where the principles of personalized medicine are not merely an adjunct but the absolute foundation of its clinical application.

10.2 Unresolved Questions and Directions for Future Research

Despite its established role, several areas warrant further investigation to optimize the use of Eliglustat and explore its broader potential.

  • Pediatric Use: While approved for use in older children in Europe, more extensive data are needed to establish its long-term safety and efficacy in the pediatric population, particularly in younger children and in those who are treatment-naïve.[13]
  • Novel Therapeutic Indications: The preclinical discovery of Eliglustat's function as an autophagy inhibitor in osteoclasts is a compelling finding.[27] This warrants clinical investigation into its potential for therapeutic repurposing in conditions where autophagy or bone resorption is pathological, such as in certain cancers, osteoporosis, or other metabolic bone diseases.
  • Management of Complex Polypharmacy: The current prescribing guidelines provide a framework for managing interactions with single CYP inhibitors. However, real-world data is needed to guide the management of patients with multiple co-morbidities and complex polypharmacy, particularly those taking multiple weak or moderate inhibitors, a scenario that is not explicitly detailed in the label.[22]
  • Long-Term Outcomes and Safety: As an orphan drug for a chronic condition, continued long-term monitoring and real-world evidence generation are essential. This will be critical for confirming its long-term safety profile, especially concerning cardiovascular health and bone disease, and for fully understanding its comparative effectiveness against ERT over decades of treatment.

Works cited

  1. Eliglustat - Wikipedia, accessed September 10, 2025, https://en.wikipedia.org/wiki/Eliglustat
  2. www.cerdelga.com, accessed September 10, 2025, https://www.cerdelga.com/#:~:text=Cerdelga%C2%AE%20(eliglustat)%20Official%20Patient%20Site
  3. Eliglustat (oral route) - Side effects & dosage - Mayo Clinic, accessed September 10, 2025, https://www.mayoclinic.org/drugs-supplements/eliglustat-oral-route/description/drg-20122508
  4. Eliglustat - brand name list from Drugs.com, accessed September 10, 2025, https://www.drugs.com/ingredient/eliglustat.html
  5. Cerdelga (eliglustat) FDA Approval History - Drugs.com, accessed September 10, 2025, https://www.drugs.com/history/cerdelga.html
  6. 205494Orig1s000 | FDA, accessed September 10, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/205494Orig1s000Approv.pdf
  7. Eliglustat | Glucosylceramide Synthase inhibitor | 99.8%(HPLC) | In Stock, accessed September 10, 2025, https://www.selleckchem.com/products/eliglustat.html
  8. Eliglustat free base | CAS#491833-29-5 | 928659-70-5 | glucosylceramide synthase, accessed September 10, 2025, https://www.medkoo.com/products/6474
  9. Eliglustat | 491833-29-5 - ChemicalBook, accessed September 10, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB52627796.htm
  10. Eliglustat | Glucosylceramide synthase inhibitor | Buy from Supplier AdooQ®, accessed September 10, 2025, https://www.adooq.com/eliglustat.html
  11. Eliglustat (Genz 99067) | Glucocerebroside Synthase Inhibitor | MedChemExpress, accessed September 10, 2025, https://www.medchemexpress.com/Eliglustat.html
  12. Cerdelga 84 mg Hard Capsules - Summary of Product Characteristics (SmPC) - (emc), accessed September 10, 2025, https://www.medicines.org.uk/emc/product/2615/smpc
  13. Cerdelga, INN-eliglustat - European Medicines Agency, accessed September 10, 2025, https://www.ema.europa.eu/en/documents/product-information/cerdelga-epar-product-information_en.pdf
  14. N-((1R,2R)-2-(2,3-Dihydro-1,4-benzodioxin-6-yl)-2-hydroxy-1-(1-pyrrolidinylmethyl)ethyl)octanamide | C23H36N2O4 | CID 23652731 - PubChem, accessed September 10, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Eliglustat
  15. Eliglustat Therapy and CYP2D6 Genotype - Medical Genetics Summaries - NCBI Bookshelf, accessed September 10, 2025, https://www.ncbi.nlm.nih.gov/books/NBK565950/
  16. Eliglustat tartrate for the treatment of adults with type 1 Gaucher ..., accessed September 10, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4554398/
  17. What is the mechanism of Eliglustat? - Patsnap Synapse, accessed September 10, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-eliglustat
  18. Eliglustat ≥98% (HPLC) - Sigma-Aldrich, accessed September 10, 2025, https://www.sigmaaldrich.com/US/en/product/sigma/sml3212
  19. Cerdelga | European Medicines Agency (EMA), accessed September 10, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/cerdelga
  20. Search Orphan Drug Designations and Approvals - FDA, accessed September 10, 2025, https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=265408
  21. highlights of prescribing information - Sanofi U.S., accessed September 10, 2025, https://products.sanofi.us/cerdelga/cerdelga.pdf
  22. Physiologically‐Based Pharmacokinetic Model Development, Validation, and Application for Prediction of Eliglustat Drug–Drug Interactions, accessed September 10, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9828395/
  23. Drugs@FDA: Drug Product Cerdelga (eliglustat), Genzyme Corporation - NDA205494/SUPPL-3, 08/29/2018 - PharmGKB, accessed September 10, 2025, https://www.pharmgkb.org/literature/15101990
  24. www.ncbi.nlm.nih.gov, accessed September 10, 2025, https://www.ncbi.nlm.nih.gov/books/NBK548849/#:~:text=By%20inhibiting%20the%20pathway%2C%20lower,animal%20models%20of%20Gaucher%20disease.
  25. Eliglustat - LiverTox - NCBI Bookshelf, accessed September 10, 2025, https://www.ncbi.nlm.nih.gov/books/NBK548849/
  26. Cerdelga eliglustat Molina Clinical Policy 227, accessed September 10, 2025, https://www.molinahealthcare.com/providers/wa/medicaid/resource/PDF/cerdelga-eliglustat-mcp227.pdf
  27. The glycosphingolipid inhibitor eliglustat inhibits autophagy in osteoclasts to increase bone mass and reduce myeloma bone disease | bioRxiv, accessed September 10, 2025, https://www.biorxiv.org/content/10.1101/2021.02.05.429906v1.full-text
  28. Eliglustat (Cerdelga®) | CUH - Cambridge University Hospitals, accessed September 10, 2025, https://www.cuh.nhs.uk/patient-information/eliglustat-cerdelga/
  29. 205494Orig1s000 | FDA - accessdata.fda.gov, accessed September 10, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/205494Orig1s000PharmR.pdf
  30. eliglustat - ClinPGx, accessed September 10, 2025, https://www.clinpgx.org/chemical/PA166123486/prescribingInfo
  31. Pharmacokinetic data summary for the free base of eliglustat tartrate - ResearchGate, accessed September 10, 2025, https://www.researchgate.net/figure/Pharmacokinetic-data-summary-for-the-free-base-of-eliglustat-tartrate_tbl3_44570535
  32. Eliglustat Genotype-Directed Drug Interaction Management - Pharmacy Times, accessed September 10, 2025, https://www.pharmacytimes.com/view/eliglustat-genotypedirected-drug-interaction-management
  33. 205494Orig1s000 - accessdata.fda.gov, accessed September 10, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/205494Orig1s000MedR.pdf
  34. Eliglustat Completed Phase 3 Trials for Gaucher Disease, Type 1 Treatment - DrugBank, accessed September 10, 2025, https://go.drugbank.com/drugs/DB09039/clinical_trials?conditions=DBCOND0035324&phase=3&purpose=treatment&status=completed
  35. Cerdelga® (eliglustat) Official Patient Site, accessed September 10, 2025, https://www.cerdelga.com/
  36. Cerdelga (eliglustat) dosing, indications, interactions, adverse effects, and more, accessed September 10, 2025, https://reference.medscape.com/drug/cerdelga-eliglustat-999938
  37. Summary of Drug Interactions - Clinical Review Report: Eliglustat (Cerdelga) - NCBI, accessed September 10, 2025, https://www.ncbi.nlm.nih.gov/books/NBK534687/
  38. Discussion - Clinical Review Report: Eliglustat (Cerdelga) - NCBI Bookshelf, accessed September 10, 2025, https://www.ncbi.nlm.nih.gov/books/NBK534689/
  39. SRT's Link to Potential Drug Interactions | National Gaucher Foundation, accessed September 10, 2025, https://www.gaucherdisease.org/blog/gaucher-srt-ert-drug-interactions-national-gaucher-foundation/
  40. CERDELGATM (eliglustat) capsules, for oral use - This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed September 10, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/205494orig1s000lbl.pdf
  41. Eliglustat Uses, Side Effects & Warnings - Drugs.com, accessed September 10, 2025, https://www.drugs.com/mtm/eliglustat.html
  42. Eliglustat Monograph for Professionals - Drugs.com, accessed September 10, 2025, https://www.drugs.com/monograph/eliglustat.html
  43. Clinical Policy: Eliglustat (Cerdelga) - Ambetter Health, accessed September 10, 2025, https://www.ambetterhealth.com/content/dam/centene/Superior/policies/pharmacy-policies/CP.PHAR.153.pdf
  44. Eliglustat Interactions Checker - Drugs.com, accessed September 10, 2025, https://www.drugs.com/drug-interactions/eliglustat.html
  45. Gaucher Disease, Type 1 | DrugBank Online, accessed September 10, 2025, https://go.drugbank.com/indications/DBCOND0035324
  46. Drugs@FDA: Drug Product Cerdelga (eliglustat), Genzyme Corporation - NDA205494/SUPPL-3, 08/29/2018 - ClinPGx, accessed September 10, 2025, https://www.clinpgx.org/literature/15101990

Published at: September 10, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.