MedPath

Agalsidase alfa Advanced Drug Monograph

Published:Aug 7, 2025

Generic Name

Agalsidase alfa

Brand Names

Replagal

Drug Type

Biotech

Associated Conditions

Fabry's Disease

Agalsidase Alfa (Replagal®): A Comprehensive Monograph on an Enzyme Replacement Therapy for Fabry Disease

Section 1: Introduction to Fabry Disease and the Rationale for Enzyme Replacement Therapy

1.1 The Pathophysiology of a Lysosomal Storage Disorder

Fabry disease, also known by a variety of historical names including Anderson-Fabry disease and angiokeratoma corporis diffusum universale, is a rare, progressive, X-linked lysosomal storage disorder.[1] The disease arises from mutations in the

GLA gene, located on the X chromosome (Xq22), which encodes the lysosomal enzyme alpha-galactosidase A (α-Gal A).[2] These mutations, of which over 300 have been described, result in a deficiency or complete absence of functional α-Gal A activity.[5] The evolution of the disease's nomenclature reflects the scientific journey from early clinical observations, such as the dermatological finding of "angiokeratoma corporis diffusum," to the identification of its primary end-organ impact in the "cardiovasorenal syndrome," and finally to the elucidation of its biochemical basis as a "ceramide trihexosidase deficiency".[1] This historical progression underscores the complex, multi-systemic nature of the disorder and highlights the fundamental need for a systemic therapeutic approach.

The X-linked inheritance pattern leads to distinct clinical presentations. Hemizygous males, who inherit the single defective X chromosome, typically experience the most severe form of the disease. Heterozygous females, who have one normal and one mutated GLA gene, can have a highly variable clinical course due to random X-chromosome inactivation (lyonization), ranging from being asymptomatic carriers to having severe manifestations comparable to males.[5] The specific

GLA mutation dictates the level of residual enzyme activity, which in turn correlates with the disease phenotype. Mutations causing little to no enzyme activity lead to the "classic" severe phenotype with early-onset symptoms, whereas mutations that permit some residual enzyme function are associated with "non-classic" or "late-onset" variants, often characterized by disease primarily affecting a single organ system, such as the heart or kidneys.[5]

1.2 The Central Role of Globotriaosylceramide (Gb3) Accumulation

The central pathological mechanism of Fabry disease is the progressive, systemic accumulation of glycosphingolipids within the lysosomes of cells throughout the body.[3] In the absence of functional α-Gal A, its primary substrate, globotriaosylceramide (Gb3, also abbreviated as GL-3), cannot be catabolized.[10] This leads to the relentless buildup of Gb3 and its deacylated, cytotoxic metabolite, globotriaosylsphingosine (lyso-Gb3), in a wide array of cell types.[9]

This pathological storage occurs in the vascular endothelium, vascular smooth muscle cells, renal podocytes and tubular epithelial cells, cardiomyocytes, and neurons of the peripheral and autonomic nervous systems.[3] The accumulation of these lipids within lysosomes disrupts normal cellular function, leading to cellular hypertrophy, inflammation, fibrosis, and ultimately, cell death and organ failure.[5] Lyso-Gb3, in particular, is considered a key biomarker and pathogenic molecule, used to differentiate between classic and non-classic phenotypes and to monitor treatment effects.[9]

1.3 Clinical Manifestations and Disease Progression

The consequences of unabated Gb3 accumulation are severe, debilitating, and ultimately life-threatening, manifesting as a multi-systemic disease that progresses over a patient's lifetime.[12] The classic phenotype often presents in childhood or adolescence with characteristic symptoms such as acroparesthesias (excruciating neuropathic pain in the hands and feet), hypohidrosis (a decreased ability to sweat), and gastrointestinal disturbances like abdominal pain and diarrhea.[3]

As the disease progresses, the ongoing cellular damage leads to major organ involvement. Renal pathology begins with proteinuria and microalbuminuria and advances to progressive chronic kidney disease, often culminating in end-stage renal disease requiring dialysis or transplantation.[5] Cardiac involvement is a major source of morbidity and mortality, presenting as progressive left ventricular hypertrophy (LVH), cardiomyopathy, valvular dysfunction, and life-threatening arrhythmias.[3] The cerebrovascular system is also critically affected, with Gb3 accumulation in cerebral blood vessels increasing the risk of transient ischemic attacks (TIAs) and ischemic strokes, which often occur at a much younger age than in the general population.[3]

1.4 The Therapeutic Principle of Enzyme Replacement Therapy (ERT)

Given that Fabry disease is caused by the deficiency of a single enzyme, the most direct therapeutic strategy is to replace it. Enzyme replacement therapy (ERT) has become the cornerstone of treatment, representing the first specific therapy developed for the disease.[2] The fundamental principle of ERT is to provide a regular intravenous supply of a functional, recombinant form of the α-Gal A enzyme.[11] This exogenous enzyme is designed to be taken up by the body's cells, delivered to the lysosomes, and there catalyze the breakdown of the accumulated Gb3.[11] By clearing the stored substrate, ERT aims to halt or slow the cascade of cellular dysfunction and organ damage, thereby modifying the natural history of the disease and preventing its most severe complications.[10] The development of ERT marked a paradigm shift from purely supportive and symptomatic care to a targeted, disease-modifying intervention, building on early proof-of-concept studies from the 1970s that used purified placental enzyme and culminating in the highly purified products made possible by modern recombinant DNA technology.[4]

Section 2: Agalsidase Alfa: Biochemical Profile and Molecular Characteristics

2.1 Recombinant Protein Production and Origin

Agalsidase alfa, marketed under the brand name Replagal®, is a recombinant form of human α-galactosidase A.[2] A defining characteristic that distinguishes it from other ERTs for Fabry disease is its manufacturing process. Agalsidase alfa is produced in a continuous human fibrosarcoma cell line (HT-1080).[18] The production relies on a proprietary gene activation technology, where the endogenous, dormant

GLA gene within the human cell line is activated in situ to produce the enzyme.[6]

This choice of a human cell line for production is a critical and strategic one. It was likely a deliberate decision aimed at producing an enzyme with post-translational modifications, particularly glycosylation, that are as close as possible to the native human enzyme. The hypothesis underlying this strategy is that a more "human-like" protein would be better tolerated by the patient's immune system, resulting in lower immunogenicity. This manufacturing choice is the fundamental origin of the key biochemical and clinical differences observed between agalsidase alfa and its main competitor, agalsidase beta.

The manufacturing process itself has evolved. It originally utilized a roller bottle (RB) platform that involved animal-derived components like bovine serum. To enhance robustness, increase operational efficiency, and mitigate risks associated with animal-derived materials, the process was later transitioned to an animal component-free (AF) bioreactor system.[8]

2.2 Molecular Structure and Physicochemical Properties

2.2.1 Amino Acid Sequence

The primary amino acid sequence of agalsidase alfa is identical to that of the native, wild-type human α-Gal A enzyme.[20] The mature, functional enzyme is a glycoprotein that exists as a homodimer, meaning it is composed of two identical protein subunits.[4] Each monomer consists of 398 amino acids, with a molecular weight of approximately 50 kDa.[19] The overall average weight of the protein is reported as 45351.6 Da, with a chemical formula of

C2029​H3080​N544​O587​S27​.[10]

2.2.2 Enzymatic Function

As a lysosomal hydrolase, agalsidase alfa belongs to the glycoside hydrolase family.[4] Its specific function is to act as a metabolizer, catalyzing the hydrolytic cleavage of terminal α-galactosyl residues from a variety of glycolipids and glycoproteins.[4] Its most clinically relevant substrate is globotriaosylceramide (Gb3).[10] The catalytic reaction proceeds via a double displacement mechanism, resulting in a net retention of the anomeric configuration.[4]

2.3 Post-Translational Modification: The Critical Role of Glycosylation

The function and fate of a recombinant protein like agalsidase alfa are determined not just by its amino acid sequence but critically by its post-translational modifications, especially N-linked glycosylation. This process, which occurs in the endoplasmic reticulum and Golgi apparatus, attaches complex carbohydrate chains (glycans) to specific asparagine residues on the protein.[6]

2.3.1 N-linked Glycosylation

Each monomer of α-Gal A has three potential sites for N-linked glycosylation, at asparagine residues N139, N192, and N215.[6] The specific structure of the glycans attached at these sites is crucial for the protein's proper folding, stability, three-dimensional structure, biodistribution, and ultimately, its functional activity and immunogenic potential.[6]

2.3.2 Mannose-6-Phosphate (M6P) Targeting

A key step in the glycosylation process is the addition of mannose-6-phosphate (M6P) residues to the oligosaccharide chains in the cis-Golgi network.[4] This M6P tag serves as a specific molecular address label. It is recognized by cation-dependent mannose-6-phosphate receptors (M6PRs) located on cell surfaces and within the trans-Golgi network.[4] This receptor-ligand interaction is the primary mechanism by which ERTs are endocytosed from the bloodstream and trafficked to the lysosomes, their site of action.[4] The efficiency of this targeting is therefore directly dependent on the M6P content of the enzyme.

2.3.3 Glycosylation Differences vs. Agalsidase Beta

While both agalsidase alfa and agalsidase beta share an identical amino acid sequence, their production in different cell lines (human vs. Chinese Hamster Ovary) results in distinct, cell-specific glycosylation patterns.[3] Analysis has shown that agalsidase alfa contains higher levels of certain monosaccharides like fucose, galactose, and N-acetylglucosamine.[4] Conversely, agalsidase beta, produced in CHO cells, is reported to have a higher degree of sialylation and, critically, a higher molar content of the M6P targeting moiety.[4]

This subtle difference in M6P content may have profound clinical implications. A lower M6P content in agalsidase alfa could translate to less efficient binding to the M6PR and consequently, less efficient delivery to the lysosome. This may be a key factor underlying the dose disparity between the two drugs. While the human cell line production of agalsidase alfa successfully yields a product with lower immunogenicity, it may come at the cost of a less optimized M6P profile for lysosomal targeting. The five-fold higher dose of agalsidase beta may be necessary to overcome this, effectively saturating the uptake pathway to achieve its observed superior biochemical effect on surrogate markers. This reveals a complex trade-off in biopharmaceutical design between optimizing for safety (immunogenicity) and optimizing for targeted delivery and potency.

Section 3: Comprehensive Pharmacological Profile

3.1 Pharmacodynamics (Mechanism of Action)

The pharmacodynamic effect of agalsidase alfa is centered on the principle of restoring a deficient metabolic pathway through enzyme replacement. The mechanism can be understood as a multi-step process.

3.1.1 Enzyme Replacement and Cellular Uptake

Agalsidase alfa is administered via intravenous infusion, introducing the recombinant enzyme into the systemic circulation.[18] From the bloodstream, it must be delivered to the lysosomes of target cells throughout the body. This crucial step is mediated by the cation-dependent mannose-6-phosphate receptor (M6PR).[7] The M6P moieties on the glycans of agalsidase alfa bind with high affinity to these receptors on the surface of cells such as vascular endothelial cells, smooth muscle cells, and renal podocytes. This binding triggers receptor-mediated endocytosis, a process where the cell membrane engulfs the enzyme-receptor complex, internalizing it into vesicles.[4]

3.1.2 Lysosomal Function Restoration

Once inside the cell, the endocytic vesicles containing the agalsidase alfa-M6PR complex are trafficked along the endosomal pathway. As these vesicles mature into late endosomes and eventually fuse with lysosomes, the internal environment becomes progressively more acidic.[4] The drop in pH to approximately 4.5–5.0 within the lysosome serves two purposes: it causes the dissociation of agalsidase alfa from its receptor, releasing the active enzyme into the lysosomal lumen, and it provides the optimal acidic environment for the enzyme to become catalytically active.[4] The M6PR is then recycled back to the cell surface to capture more enzyme.

3.1.3 Substrate Hydrolysis

Within the lysosome, the now-active agalsidase alfa performs its intended function. It acts as a metabolizer, specifically targeting and hydrolyzing the terminal α-galactosyl bond on its primary substrate, globotriaosylceramide (Gb3), as well as other related glycosphingolipids.[4] This enzymatic action breaks down the accumulated lipids into smaller molecules that can be further metabolized or cleared by the cell. By restoring this catabolic pathway, agalsidase alfa reduces the lysosomal burden of Gb3, which is the intended mechanism to halt the pathophysiology of Fabry disease and prevent or mitigate its severe clinical manifestations, including renal failure, cardiomyopathy, and cerebrovascular events.[10]

3.2 Pharmacokinetics

The pharmacokinetic profile of agalsidase alfa describes its movement into, through, and out of the body, which dictates its dosing regimen and highlights key characteristics of this class of therapy.

3.2.1 Absorption and Distribution

As a protein therapeutic administered intravenously, absorption is immediate and complete. Following a standard 0.2 mg/kg infusion, agalsidase alfa exhibits a biphasic serum distribution and elimination profile.[15] In patients without end-stage renal disease, the peak plasma concentration (

Cmax​) reaches approximately 3710±855 U/mL, with a total drug exposure (Area Under the Curve, AUC) of 256,958±63,499 min*U/mL.[10] The volume of distribution at steady state (

Vd,ss​) is about 17% of body weight, indicating that the drug is distributed primarily in the plasma and extracellular fluid compartments, with limited penetration into deeper tissues from the circulation alone.[10] As a large glycoprotein, it is not expected to be bound to other plasma proteins in circulation.[10]

3.2.2 Metabolism and Elimination

Agalsidase alfa is a protein and is therefore expected to be metabolized through the same pathways as other endogenous proteins. Its degradation occurs via nonspecific proteolysis, where proteases and other enzymes break it down into smaller peptides and constituent amino acids.[10] These amino acids then enter the body's general amino acid pool and can be reused for the synthesis of new proteins or further catabolized and eliminated by the kidneys.[10] The drug is not metabolized by the cytochrome P450 (CYP450) enzyme system in the liver, making CYP450-mediated drug-drug interactions highly unlikely.[18]

3.2.3 Half-life and Clearance

The elimination half-life (t1/2​) of agalsidase alfa is relatively short, reported to be 108±17 minutes for males and 89±28 minutes for females.[10] Systemic clearance is approximately 2.66 mL/min/kg for males and 2.10 mL/min/kg for females.[10] Studies have also noted that clearance is faster in pediatric patients compared to adults.[15]

This pharmacokinetic profile, particularly the short half-life of roughly 90-110 minutes, is a defining feature of first-generation ERTs. The rapid clearance from the circulation means that to maintain therapeutically relevant enzyme levels in target tissues, the drug must be administered frequently. This directly explains the necessity of the chronic, bi-weekly infusion schedule that is a significant treatment burden for patients.[18] This limitation is not merely a data point but the primary clinical and commercial driver for innovation in the field. The development of next-generation therapies for Fabry disease, such as the PEGylated enzyme pegunigalsidase alfa, was undertaken specifically to engineer a molecule with a much longer half-life, aiming to reduce infusion frequency and improve patient quality of life.[17] This connection demonstrates how the fundamental pharmacology of agalsidase alfa has directly shaped the entire trajectory of subsequent drug development for Fabry disease.

Section 4: Clinical Development and Efficacy Analysis

The clinical efficacy of agalsidase alfa has been established through a comprehensive development program that includes pivotal placebo-controlled trials, long-term open-label extensions, and extensive real-world data from observational registries. This body of evidence demonstrates its role in modifying the natural history of Fabry disease across various domains and patient populations.

Table 1: Summary of Key Clinical Trials for Agalsidase Alfa

Trial Identifier/NamePhaseDesignPatient Population (N)Dosage RegimenPrimary Endpoint(s)Key Efficacy ResultsSource(s)
Schiffmann et al. (2001)Phase II/IIIRandomized, Double-Blind, Placebo-Controlled26 adult males0.2 mg/kg EOWChange in Brief Pain Inventory (BPI) 'pain at its worst' scoreStatistically significant reduction in neuropathic pain vs. placebo.14
Moore et al. (2001)Phase II/IIIRandomized, Double-Blind, Placebo-Controlled15 adult males0.2 mg/kg EOWChange in left ventricular mass (LVM) indexSignificant reduction in LVM index vs. placebo, which showed an increase.20
HGT-REP-084 (NCT01363492)Phase IIOpen-Label, Multicenter14 ERT-naïve children (≥7 years)0.2 mg/kg EOWSafety, changes in autonomic functionWell tolerated; no deterioration in cardiac or renal function over 55 weeks; suggested benefit of early ERT initiation.29
FOS (Fabry Outcome Survey) (NCT03289065)Observational RegistryInternational, Observational>2,100 patients (all ages/sexes)0.2 mg/kg EOW (standard)Long-term safety and effectivenessSlowed eGFR decline, stabilized cardiomyopathy, and delayed morbidity/mortality compared to untreated historical cohorts over up to 20 years.13
Japanese PMSPost-Marketing SurveillanceAll-case surveillance493 adult patients0.2 mg/kg EOWLong-term safety and effectivenessConfirmed long-term tolerability and control of renal and cardiac symptom progression over a mean of 3.5-9.6 years.31

4.1 Pivotal Placebo-Controlled Trials in Adult Males

The foundational evidence for the efficacy of agalsidase alfa was generated in two landmark randomized, double-blind, placebo-controlled trials conducted in adult male patients with Fabry disease.[20] These 6-month studies provided the first robust demonstration of its clinical benefits.

In a trial involving 26 patients, the primary endpoint was neuropathic pain, a debilitating early symptom of the disease. Treatment with agalsidase alfa at a dose of 0.2 mg/kg every other week (EOW) resulted in a statistically and clinically significant reduction in pain scores as measured by the Brief Pain Inventory (BPI), compared to patients receiving placebo.[14]

In a parallel trial with 15 patients, the focus was on cardiac involvement. The primary endpoint was the change in left ventricular mass (LVM) index, a key indicator of cardiac hypertrophy driven by Gb3 accumulation. After 6 months, patients treated with agalsidase alfa showed a significant reduction in their LVM index, whereas patients in the placebo group experienced a continued increase in cardiac mass.[20] These trials also showed a significant improvement in creatinine clearance in the treated group, although another measure of renal function, inulin clearance, was not significantly different from placebo.[20]

4.2 Long-Term Efficacy and Real-World Evidence

While short-term placebo-controlled trials are essential for regulatory approval, the chronic, progressive nature of Fabry disease necessitates long-term data to assess true disease-modifying effects.

4.2.1 Open-Label Extension (OLE) Studies

Patients who completed the initial pivotal trials were eligible to enroll in open-label extension studies, where all participants received agalsidase alfa. These extensions demonstrated that the clinical benefits observed in the first 6 months were sustained over longer periods. Reductions in LVM and improvements in pain scores were maintained with continued therapy, with some benefits, such as improvements in sweat function and thermal sensation thresholds, becoming apparent only after several years of treatment.[20]

4.2.2 The Fabry Outcome Survey (FOS)

The most extensive evidence for the long-term effectiveness of agalsidase alfa comes from the Fabry Outcome Survey (FOS), an international, multicenter, observational registry initiated in 2001.[15] With data from over 2,100 patients spanning up to two decades, FOS provides invaluable real-world insights into the impact of treatment.[13] Analyses of FOS data, when compared with the known natural history of untreated Fabry disease, have shown that long-term treatment with agalsidase alfa provides substantial clinical benefits. These include a slowing of the rate of decline in renal function (estimated glomerular filtration rate, or eGFR), stabilization or slowing of the progression of cardiomyopathy, and a delay in overall morbidity and mortality.[13] A parallel long-term post-marketing surveillance study in Japan, conducted over 8-10 years, corroborated these findings, demonstrating sustained reductions in the biomarker Gb3 and effective control over the progression of renal and cardiac symptoms in a real-world setting.[31]

4.3 Efficacy in Specific Populations

The clinical development program also included dedicated studies and sub-analyses to evaluate efficacy in key patient populations beyond adult males.

4.3.1 Female Patients

Although Fabry disease is X-linked, heterozygous females can experience significant disease burden. Studies and analyses from the FOS registry have shown that female patients treated with agalsidase alfa experience benefits comparable to those seen in males. This includes significant reductions in left ventricular mass and stabilization of disease progression, confirming its efficacy in this often-undertreated population.[15]

4.3.2 Pediatric Patients

Treating children with Fabry disease is critical to preventing the onset of irreversible organ damage. An open-label, multicenter Phase II study (NCT01363492) evaluated agalsidase alfa in 14 ERT-naïve children aged 7 years and older.[29] Over 55 weeks of treatment, the therapy was found to be well tolerated. As expected in a relatively short trial in a patient population with less advanced disease, there were no significant changes in key efficacy endpoints like LVMI or eGFR.[29] However, the key conclusion from this and other pediatric studies is that initiating ERT early in life, before the establishment of significant organ pathology, is crucial for maximizing long-term clinical benefit and potentially slowing or preventing disease progression.[18]

4.3.3 Patients with Advanced Renal Disease

The collective body of clinical evidence reveals a critical principle of ERT: its efficacy is greatest when initiated before the development of advanced, irreversible organ damage. In patients who already have extensive renal damage and fibrosis, the ability of ERT to improve or even stabilize kidney function is limited.[18] Long-term follow-up data have shown that patients who began therapy with pre-existing stage 3 chronic kidney disease continued to experience a decline in eGFR.[20] While the rate of this decline appeared to be slower than that observed in historical untreated cohorts, it underscores that ERT cannot reverse established fibrosis. This finding transforms the efficacy data from a simple list of outcomes into a powerful clinical imperative. It strongly advocates for early diagnosis, ideally through newborn screening programs, and prompt initiation of therapy in younger, less symptomatic patients to prevent the progression to later stages of the disease where treatment is far less effective.

Section 5: Safety, Tolerability, and Immunogenicity

The safety profile of agalsidase alfa has been extensively characterized through clinical trials and over two decades of post-marketing surveillance. It is generally considered to be well-tolerated, with a predictable and manageable side effect profile.

Table 2: Comprehensive Safety Profile of Agalsidase Alfa

CategorySpecific Events/ParametersIncidence/FrequencyClinical Notes/ManagementSource(s)
Infusion-Associated Reactions (IARs)Rigors, pyrexia (fever), flushing, headache, nausea, chills, tremor, dyspnea, pruritusOccur in ~13–24% of patients.Most common adverse events; typically mild-to-moderate and transient. Often occur at the start of therapy and diminish over time. Can be managed with premedication (antihistamines, corticosteroids).14
Common Adverse EventsIn addition to IARs: fatigue/lethargy, abdominal pain, diarrhea, dizziness, muscle pain, hypertension, sore throat.Variable, generally mild.Consistent with the known safety profile and underlying symptoms of Fabry disease.33
Serious Adverse EventsSevere hypersensitivity/anaphylactic reactions.Rare.If severe reactions occur, infusion should be stopped immediately and appropriate emergency treatment initiated.14
ImmunogenicityIgG Antibody Formation: Low-titer anti-drug antibodies (ADAs).Observed in ~24% of male patients, typically within 3-12 months. Not reported in female patients.No clinically significant effect on safety or efficacy observed in trials.14
Immunologic Tolerance: Disappearance of IgG antibodies over time.Develops in a subset of patients (~7% of total cohort after 12-54 months).Suggests the immune system can adapt to the therapy over time.18
IgE Antibody Status: Antibodies associated with Type I hypersensitivity.No IgE antibodies have been detected in any patient.This is a key safety differentiator, indicating a very low risk of true allergic/anaphylactic reactions.14
ContraindicationsHypersensitivityAbsolute contraindication.Patients with a known hypersensitivity to agalsidase alfa or its excipients should not receive the drug.18
Serious Drug InteractionsInhibitors of intracellular α-galactosidase activity.Amiodarone, Chloroquine, Benoquin (Monobenzone), Gentamicin.Co-administration is contraindicated or strongly cautioned against as these drugs can theoretically inhibit the therapeutic enzyme's activity within the lysosome.10
Pharmacokinetic EnhancersMigalastatThe chaperone therapy migalastat can increase the serum concentration of agalsidase alfa.10

5.1 Adverse Event Profile and Tolerability

5.1.1 Infusion-Associated Reactions (IARs)

The most frequently reported adverse events associated with agalsidase alfa are infusion-associated reactions (IARs).[14] These events, which occur during or shortly after the intravenous infusion, are reported in approximately 13% to 24% of patients.[14] The characteristic symptoms are generally mild to moderate in severity and include rigors (shaking chills), pyrexia (fever), flushing, headache, nausea, and pruritus (itching).[18] These reactions are most common during the initial phase of treatment and tend to decrease in frequency and severity with subsequent infusions as the patient's body adapts to the therapy.[14]

5.1.2 Management and Home Infusion

The management of IARs is typically straightforward. For patients who experience them, premedication with agents such as antihistamines, antipyretics (e.g., paracetamol), and/or corticosteroids before the infusion can effectively prevent or alleviate the symptoms.[33] A key advantage for long-term management is the feasibility of home infusion. For patients who are stable and tolerate their infusions well, transitioning from a hospital or clinic setting to home-based administration is a common practice that significantly improves convenience and quality of life.[14] In the event of an adverse reaction during a home infusion, patients are instructed to stop the infusion immediately and seek medical attention.[25]

5.1.3 Serious Adverse Events

Long-term surveillance, including data from the FOS registry and dedicated post-marketing studies, has consistently shown that serious adverse events deemed to be related to agalsidase alfa are rare.[14] Studies evaluating the newer, animal component-free bioreactor-produced version of the drug have not identified any new safety signals, confirming a stable and favorable long-term safety profile.[8]

5.2 Immunogenicity

As with all therapeutic proteins, agalsidase alfa has the potential to elicit an immune response, a phenomenon known as immunogenicity.

5.2.1 IgG Antibody Formation

The most common immune response is the development of non-neutralizing, low-titer immunoglobulin G (IgG) antibodies, also known as anti-drug antibodies (ADAs). This response is observed almost exclusively in male patients, occurring in approximately 24% of this population.[18] These antibodies typically appear within the first 3 to 12 months of initiating therapy.[18] Importantly, clinical trials have not found a clinically significant impact of these low-titer IgG antibodies on the overall safety or efficacy of the treatment.[15] In stark contrast, antibody formation has not been reported in female patients receiving agalsidase alfa, likely due to the presence of some endogenous enzyme leading to immune tolerance.[14]

5.2.2 Immunologic Tolerance

Over time, a subset of male patients who initially develop IgG antibodies can go on to develop immunologic tolerance. This is characterized by a decline in antibody titers and, in some cases, the complete disappearance of detectable antibodies. One long-term analysis found that after 12 to 54 months of therapy, while 17% of the total male patient cohort remained antibody-positive, 7% had shown evidence of developing tolerance.[18]

5.2.3 Hypersensitivity and IgE Antibodies

Severe hypersensitivity or anaphylactic reactions are a potential risk with any protein infusion and have been rarely reported with agalsidase alfa.[18] However, a critical and distinguishing feature of agalsidase alfa's safety profile is the consistent finding that

no immunoglobulin E (IgE) antibodies have been detected in any treated patient.[14] IgE antibodies are the mediators of true Type I allergic reactions (anaphylaxis). Their absence strongly suggests that the immune response to agalsidase alfa is not of the classic allergic type. This favorable immunogenicity profile is a direct and predictable consequence of its production in a human cell line. The use of a human expression system results in a protein with human-like glycosylation patterns, which the patient's immune system is less likely to recognize as foreign compared to proteins with non-human glycan structures. This makes the development of a true allergic response highly unlikely and represents a core component of the drug's value proposition and a key point of differentiation from ERTs produced in non-human cell lines.

5.3 Contraindications and Drug Interactions

5.3.1 Contraindications

The only absolute contraindication for agalsidase alfa is a known history of hypersensitivity to the active substance or to any of the excipients in the formulation.[18]

5.3.2 Drug Interactions

Clinically significant drug interactions are limited but important. Agalsidase alfa should not be co-administered with substances that are known to be potent inhibitors of intracellular α-galactosidase activity. This is because such drugs could theoretically enter the lysosome and directly inhibit the therapeutic enzyme, rendering the ERT ineffective. Drugs in this category include the antiarrhythmic amiodarone, the antimalarial chloroquine, the depigmenting agent benoquin (monobenzone), and the antibiotic gentamicin.[10] Additionally, co-administration with the oral chaperone therapy

migalastat may lead to an increased serum concentration of agalsidase alfa, an interaction that may require monitoring.[10]

Section 6: Comparative Analysis: Agalsidase Alfa vs. Agalsidase Beta

The clinical and commercial story of agalsidase alfa is inextricably linked to that of its contemporary, agalsidase beta (Fabrazyme®). While both are recombinant α-Gal A enzymes intended for the same purpose, they differ in manufacturing, dosing, and certain efficacy and safety parameters. This comparison is central to understanding the therapeutic landscape of Fabry disease.

Table 3: Head-to-Head Comparison of Agalsidase Alfa (Replagal®) and Agalsidase Beta (Fabrazyme®)

AttributeAgalsidase Alfa (Replagal®)Agalsidase Beta (Fabrazyme®)Source(s)
ManufacturerShire (now Takeda)Genzyme (now Sanofi)2
Production Cell LineHuman Fibrosarcoma (HT-1080)Chinese Hamster Ovary (CHO)3
Amino Acid SequenceIdentical to native human α-Gal AIdentical to native human α-Gal A20
Key Glycosylation FeaturesHuman-like pattern; higher levels of fucose, galactose, N-acetylglucosamine.Non-human pattern; higher levels of mannose-6-phosphate (M6P) and sialic acid.4
Registered Dose (EOW)0.2 mg/kg1.0 mg/kg3
Annual Cost (approx.)~€210,000 (for 70 kg patient)~€210,000 (for 70 kg patient)37
Efficacy - Clinical EventsSimilar rate of major clinical events (renal, cardiac, cerebrovascular, death).Similar rate of major clinical events.9
Efficacy - Biochemical (lyso-Gb3)Significant reduction, but less robust.Superior reduction, especially in classic males.9
Efficacy - Cardiac (LVMI)Significant reduction.Superior reduction in LVMI after 1 year.9
Efficacy - Renal (eGFR)Similar stabilization/slope of eGFR decline.Similar stabilization/slope of eGFR decline.9
Immunogenicity (IgG/ADA)Lower risk. ~24-28% of classic males develop IgG antibodies. No IgE detected.Higher risk. ~52% of classic males develop IgG antibodies. IgE-mediated reactions reported, though rare.9
Regulatory StatusEMA approved (Europe, Canada, Australia, etc.). Not FDA approved.EMA and FDA approved.2

6.1 The Dosing and Cost Conundrum

The most striking difference between the two therapies is the five-fold disparity in their approved bi-weekly doses: 0.2 mg/kg for agalsidase alfa versus 1.0 mg/kg for agalsidase beta.[3] This difference is not reflected in their pricing. In European markets, the annual cost of therapy for a standard 70 kg patient is remarkably similar, at approximately €210,000 for both drugs.[37] This means that on a per-milligram basis, agalsidase alfa is about five times more expensive than agalsidase beta, a fact that has significant implications for healthcare payers and formulary decisions.

6.2 Comparative Efficacy: Clinical vs. Biomarker Outcomes

The dose difference naturally leads to the question of comparative efficacy. A substantial body of evidence, including retrospective cohort studies and at least one direct head-to-head trial, has compared the two enzymes at their licensed doses, revealing a nuanced and complex picture.

6.2.1 Major Clinical Events

When assessing hard clinical endpoints—the composite of major renal events (e.g., end-stage renal disease), cardiac events (e.g., myocardial infarction), cerebrovascular events (e.g., stroke), and death—the evidence consistently shows no significant difference in the event rate between patients treated with agalsidase alfa and those treated with agalsidase beta.[9] Propensity score-matched analyses have confirmed this finding, suggesting that from the perspective of preventing major, life-altering complications, the two therapies perform comparably over the long term.

6.2.2 Biochemical and Surrogate Markers

In stark contrast to the clinical event data, studies consistently demonstrate that the higher 1.0 mg/kg dose of agalsidase beta leads to a more robust and superior effect on key biomarkers and surrogate endpoints. Specifically, agalsidase beta achieves a significantly greater reduction in plasma levels of the biomarker lyso-Gb3, an effect that is most pronounced in men with the classic Fabry phenotype.[9] Furthermore, agalsidase beta has been shown to produce a better reduction in left ventricular mass index (LVMI) after the first year of treatment.[9] On measures of renal function, however, the two drugs appear to perform similarly, with comparable rates of eGFR decline over time.[9]

This disconnect between biomarker/surrogate outcomes and hard clinical outcomes is a central paradox in the comparison of these two drugs. It raises fundamental questions about drug development and clinical practice. One interpretation is that there may be a "threshold effect," where the 0.2 mg/kg dose of agalsidase alfa is sufficient to achieve the necessary level of Gb3 clearance to prevent the majority of major clinical events. In this model, the additional biochemical clearance provided by the higher dose of agalsidase beta may provide diminishing returns in terms of preventing hard clinical outcomes, even while it improves surrogate markers. Alternatively, it is possible that the surrogate markers, like lyso-Gb3 reduction, are not perfectly correlated with long-term clinical benefit, or that the studies conducted to date have been underpowered to detect a true, albeit smaller, difference in clinical event rates.[36] This reframes the debate from a simple question of "which drug is better?" to a more sophisticated discussion about dosing philosophy, the clinical relevance of surrogate endpoints, and the cost-effectiveness of pursuing maximal biochemical correction if it does not translate into a demonstrable difference in preventing strokes or kidney failure.

6.3 Comparative Immunogenicity

The different manufacturing platforms lead to a clear difference in immunogenicity. The risk of developing IgG anti-drug antibodies is significantly higher for patients treated with agalsidase beta, with an odds ratio of approximately 2.8 compared to agalsidase alfa.[9] In men with classic Fabry disease, persisting antibodies are found in over 50% of those treated with agalsidase beta, compared to under 30% of those on agalsidase alfa.[36]

Interestingly, the clinical impact of these antibodies appears to differ. In patients on agalsidase alfa, the presence of antibodies is associated with a less pronounced reduction in lyso-Gb3. However, for patients on agalsidase beta, the five-fold higher dose appears to be sufficient to overcome the inhibitory effect of the antibodies, leading to a robust biochemical response even in antibody-positive patients.[36] This suggests that while agalsidase alfa offers a better safety profile from an immunogenicity standpoint, agalsidase beta's high-dose strategy may compensate for its higher immunogenic potential.

Section 7: Regulatory and Market Access Trajectory

The regulatory history of agalsidase alfa is a tale of two continents, marked by early success in Europe and a complex, ultimately unsuccessful, journey in the United States. This history has profoundly shaped the competitive landscape for Fabry disease therapies.

7.1 European Union and Global Approval

Agalsidase alfa, as Replagal®, secured marketing authorization from the European Medicines Agency (EMA) on August 3, 2001, becoming one of the first specific treatments for Fabry disease.[2] Due to the rarity of the disease and the limited data available at the time, it was initially granted authorization under 'exceptional circumstances'.[26] This status is used when a comprehensive data package is not feasible at the time of approval but the product addresses an unmet need. The 'exceptional circumstances' designation was lifted on July 20, 2015, after the manufacturer, Shire, provided the additional long-term data required by the agency.[26] Following its European debut, Replagal gained approval in many other jurisdictions, including Canada, Australia, and Japan, establishing it as a standard of care for Fabry disease globally.[3]

7.2 The United States FDA Experience: A Case Study in Regulatory Hurdles

In stark contrast to its global success, agalsidase alfa has never been approved for use in the United States.[2] Its path to the U.S. market is a compelling case study in the interplay of regulatory standards, market dynamics, and corporate strategy.

7.2.1 Initial Encouragement and BLA Submission

The opportunity for U.S. market entry arose under unusual circumstances. In 2009, Genzyme (the manufacturer of agalsidase beta, marketed as Fabrazyme®) experienced significant manufacturing problems, leading to a severe and prolonged global shortage of the only FDA-approved ERT for Fabry disease.[44] Facing a critical unmet need for U.S. patients, the Food and Drug Administration (FDA) took the proactive step of encouraging Shire to submit a Biologics License Application (BLA) for Replagal in both 2009 and 2011, with the goal of providing a much-needed therapeutic alternative.[45] Shire complied, submitting its BLA in late 2009 and providing the drug to some U.S. patients through an emergency access program.[41]

7.2.2 Withdrawal of the BLA

Hopes for a swift approval were dashed. In March 2012, Shire announced the surprising decision to withdraw its BLA for Replagal.[45] The company's official statement clarified that recent interactions with the FDA had led them to believe that the agency would require

additional, new controlled clinical trials to support an approval.[45] Shire concluded that conducting these trials would be a lengthy and costly process, delaying a potential approval until the "distant future," and therefore made the strategic decision to withdraw the application.[47] The FDA did not raise any new concerns regarding the safety profile of Replagal during this process.[46]

7.2.3 The Shifting Landscape

Shire's decision was undoubtedly influenced by a rapidly changing landscape. By 2012, Genzyme was making significant progress in resolving its manufacturing issues and restoring the full supply of Fabrazyme, diminishing the urgency that had initially prompted the FDA's encouragement.[45] Shire likely performed a risk-reward calculation, weighing the substantial financial burden and uncertain outcome of new clinical trials against the reality of competing with an entrenched and resurgent market leader.

This withdrawal proved to be a pivotal moment. It effectively cemented a near-monopoly for Fabrazyme in the lucrative U.S. market for almost a decade and set a high regulatory bar for future entrants. The FDA's stance underscored its requirement for robust, controlled data, even in the face of supply shortages. This precedent was reinforced in 2021 when Fabrazyme's approval was converted from an accelerated to a traditional one, based on long-term data.[12] This made the regulatory path even more challenging for subsequent therapies, such as pegunigalsidase alfa, which could no longer easily use an accelerated approval pathway without demonstrating a clear and meaningful benefit over an available, traditionally-approved therapy.[12] The story of Replagal's U.S. application is therefore less about the drug's intrinsic merit and more a lesson in the powerful influence of regulatory timing, competitive context, and corporate risk assessment on market access.

Section 8: Therapeutic Context and Future Perspectives

8.1 Current Place in Therapy

In the numerous markets where it is approved, agalsidase alfa remains an established and important first-line enzyme replacement therapy for patients with a confirmed diagnosis of Fabry disease.[3] It is indicated for long-term use in adults and children aged 7 years and older. Its value is anchored in over two decades of clinical use, a favorable immunogenicity profile, and a vast body of long-term safety and efficacy data from the FOS registry that confirms its ability to positively alter the natural course of the disease.[13] In clinical practice, the choice between agalsidase alfa and agalsidase beta often hinges on factors such as physician experience and preference, regional availability, and national or hospital-level tendering processes, particularly given that large-scale studies have shown similar long-term outcomes on hard clinical events.[9]

8.2 The Evolving Therapeutic Landscape

The treatment paradigm for Fabry disease is no longer limited to the two pioneering ERTs. A wave of innovation is bringing new therapeutic modalities to the forefront, each seeking to address the limitations of first-generation enzymes. Agalsidase alfa now finds itself in a competitive landscape bracketed by therapies offering improved pharmacokinetics, greater convenience, or entirely new mechanisms of action.

8.2.1 Next-Generation ERT (Pegunigalsidase alfa)

Pegunigalsidase alfa (Elfabrio®), approved by the FDA and EMA in 2023, represents a direct evolution of ERT.[17] It is a form of agalsidase alfa that has been chemically modified through PEGylation (the attachment of polyethylene glycol chains) and is produced in a plant cell-based system.[17] This PEGylation dramatically extends the enzyme's plasma half-life from approximately 2 hours to around 80 hours.[17] This improved pharmacokinetic profile offers the potential for better tissue distribution, reduced immunogenicity, and improved tolerability. The pivotal head-to-head BALANCE trial demonstrated that pegunigalsidase alfa was non-inferior to agalsidase beta in slowing the rate of renal function decline in patients with deteriorating kidney function, establishing it as a major new competitor.[17]

8.2.2 Chaperone Therapy (Migalastat)

Migalastat (Galafold®) is an oral small molecule that represents a completely different therapeutic approach.[35] It acts as a pharmacological chaperone. Instead of replacing the enzyme, migalastat binds to and stabilizes certain unstable, mutant forms of the α-Gal A enzyme within the cell. This stabilization helps the misfolded enzyme to traffic correctly to the lysosome and perform its function.[10] The major advantage of migalastat is its oral route of administration, which eliminates the burden of intravenous infusions. However, its significant limitation is that it is only effective for patients who have

GLA mutations that are "amenable" to chaperoning, which constitutes a specific subset of the Fabry patient population.[17]

8.2.3 Substrate Reduction Therapy (SRT)

Another emerging class of oral medications is substrate reduction therapy (SRT). Investigational drugs like lucerastat and venglustat work by inhibiting an upstream enzyme in the glycosphingolipid synthesis pathway, specifically glucosylceramide synthase.[27] By blocking this enzyme, SRTs reduce the rate at which Gb3 is produced in the first place, thereby lowering the amount of substrate that accumulates in the lysosomes.[61] This mechanism is independent of the patient's own α-Gal A activity and offers the potential to cross the blood-brain barrier, which could allow for the treatment of the neurological manifestations of Fabry disease that are not well-addressed by ERTs.[27]

8.2.4 Gene Therapy

The ultimate therapeutic goal for monogenic disorders like Fabry disease is a one-time, potentially curative treatment. Gene therapy is the most promising strategy to achieve this. Several approaches are currently in clinical development, including both in vivo and ex vivo methods.[27]

In vivo strategies typically use adeno-associated virus (AAV) vectors to deliver a functional copy of the GLA gene directly to the patient's cells (often liver cells). Ex vivo strategies involve harvesting a patient's own hematopoietic stem cells, using a lentiviral vector to insert the correct GLA gene into them in a lab, and then re-infusing these corrected cells back into the patient.[27] Both approaches aim to create a continuous, endogenous source of the α-Gal A enzyme, potentially freeing patients from the need for lifelong chronic therapy.

This rapidly advancing landscape positions agalsidase alfa as a foundational, legacy product. It is being challenged on pharmacokinetic grounds by next-generation ERTs, on convenience by oral therapies, and on long-term vision by gene therapy. Its future role will likely be defined by its extensive long-term safety record, patient and physician loyalty, and competitive pricing, but it is clear that it is a product of a previous, albeit pioneering, era of therapeutic design for Fabry disease.

Section 9: Expert Synthesis and Conclusion

9.1 Summary of Agalsidase Alfa's Profile

Agalsidase alfa (Replagal®) is a well-established enzyme replacement therapy that has been a cornerstone of Fabry disease management for over two decades in many parts of the world. As a recombinant version of the human α-galactosidase A enzyme, its primary mechanism is to restore the metabolic function that is lost in this devastating lysosomal storage disorder. Its development and approval marked a significant milestone, shifting the treatment paradigm from purely symptomatic care to a targeted, disease-modifying strategy. The strengths of agalsidase alfa are rooted in its unique manufacturing process and the extensive body of evidence supporting its long-term use. Produced in a human cell line, it possesses a favorable immunogenicity profile, notably characterized by a lower incidence of anti-drug antibodies compared to its main competitor and a complete absence of IgE-mediated responses. Furthermore, vast real-world data from the Fabry Outcome Survey (FOS) have confirmed its long-term clinical benefit in slowing the progression of renal and cardiac disease and improving survival compared to the natural history of untreated patients.

9.2 Critical Assessment of Strengths and Limitations

A balanced, expert assessment of agalsidase alfa reveals a profile of clear strengths counterweighed by significant limitations that have defined its trajectory and current standing.

Strengths:

  • Proven Long-Term Benefit: Decades of data from clinical trials and large-scale observational registries provide robust evidence that agalsidase alfa modifies the natural history of Fabry disease, stabilizing organ function and delaying major clinical events.
  • Favorable Immunogenicity Profile: Its production in a human cell line confers a distinct safety advantage, with a lower risk of IgG antibody formation and no reported cases of IgE antibody development, minimizing the risk of true allergic reactions.
  • Established Safety and Tolerability: The therapy is generally well-tolerated, with predictable and manageable infusion-associated reactions. Its established safety profile supports its use in a home-infusion setting, enhancing patient quality of life.

Limitations:

  • Lack of FDA Approval: The failure to secure approval in the United States represents its single greatest commercial and clinical limitation, severely restricting its market reach and leaving U.S. patients with fewer therapeutic options for many years.
  • Sub-maximal Biomarker Response: The registered dose of 0.2 mg/kg EOW, while demonstrating comparable efficacy on hard clinical endpoints, achieves a less robust reduction in key biochemical (lyso-Gb3) and cardiac surrogate (LVMI) markers compared to the higher dose of agalsidase beta. This creates clinical ambiguity regarding optimal dosing and the long-term significance of surrogate marker normalization.
  • Inherent Treatment Burden: As a first-generation ERT with a short half-life, it requires lifelong, bi-weekly intravenous infusions, which constitutes a significant and cumulative burden on patients and healthcare systems.

9.3 Concluding Remarks: An Enduring but Evolving Role

Agalsidase alfa stands as a foundational and pioneering therapy in the treatment of Fabry disease. Its history offers a compelling narrative on the intricate interplay between innovative biochemical design, rigorous clinical development, complex regulatory hurdles, and dynamic market forces. While it remains a vital and effective therapeutic option for thousands of patients in Europe and other regions, its position in the therapeutic algorithm is being fundamentally challenged by a new generation of innovation.

The future of Fabry disease management is moving towards a more personalized and convenient model. Agalsidase alfa is now flanked by next-generation ERTs with superior pharmacokinetics, targeted oral chaperone therapies for specific patient subsets, broadly applicable oral substrate reduction therapies, and the ultimate promise of one-time curative gene therapies. In this evolving landscape, the role of first-generation enzymes like agalsidase alfa will inevitably be redefined. It will likely endure as a reliable option, valued for its unparalleled long-term safety data and established track record, but its market share and position as a first-choice therapy will face increasing pressure. The journey of agalsidase alfa, from a breakthrough therapy to an established incumbent, serves as a powerful illustration of the relentless cycle of innovation in biotechnology.

Works cited

  1. Fabry Disease Recruiting Phase 3 Trials for Agalsidase alfa (DB15874) | DrugBank Online, accessed August 7, 2025, https://go.drugbank.com/indications/DBCOND0000667/clinical_trials/DB15874?phase=3&status=recruiting
  2. What is Agalsidase alfa used for? - Patsnap Synapse, accessed August 7, 2025, https://synapse.patsnap.com/article/what-is-agalsidase-alfa-used-for
  3. Agalsidase alfa – Knowledge and References - Taylor & Francis, accessed August 7, 2025, https://taylorandfrancis.com/knowledge/Medicine_and_healthcare/Physiology/Agalsidase_alfa/
  4. Agalsidase alfa for the treatment of fabry disease: a closer look, accessed August 7, 2025, https://www.openaccessjournals.com/articles/agalsidase-alfa-for-the-treatment-of-fabry-disease-a-closer-look.pdf
  5. Agalsidase alfa: enzyme therapy for Anderson–Fabry disease - Open Access Journals, accessed August 7, 2025, https://www.openaccessjournals.com/articles/agalsidase-alfa-enzyme-therapy-for-anderson-fabry-disease.pdf
  6. Importance of glycosylation in enzyme replacement therapy - Fabry ..., accessed August 7, 2025, https://www.ncbi.nlm.nih.gov/books/NBK11598/
  7. Agalsidase alfa (Replagal™) in the treatment of Anderson-Fabry disease - PMC, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2721310/
  8. The Safety of Agalsidase Alfa Enzyme Replacement Therapy in Canadian Patients with Fabry Disease Following Implementation of a Bioreactor Process - PMC - PubMed Central, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8602602/
  9. Agalsidase alfa versus agalsidase beta for the treatment of Fabry disease: an international cohort study | Journal of Medical Genetics, accessed August 7, 2025, https://jmg.bmj.com/content/55/5/351
  10. Agalsidase alfa: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed August 7, 2025, https://go.drugbank.com/drugs/DB15874
  11. What is the mechanism of Agalsidase alfa? - Patsnap Synapse, accessed August 7, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-agalsidase-alfa
  12. 761161Orig1s000 - accessdata.fda.gov, accessed August 7, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2023/761161Orig1s000MultidisciplineR.pdf
  13. Long-term agalsidase alfa in Fabry disease shows treatment benefit, accessed August 7, 2025, https://fabrydiseasenews.com/news/treatment-benefit-long-term-agalsidase-alfa-fabry-disease/
  14. Safety of enzyme replacement therapy - Fabry Disease - NCBI Bookshelf, accessed August 7, 2025, https://www.ncbi.nlm.nih.gov/books/NBK11617/
  15. Update on role of agalsidase alfa in management of Fabry disease - PMC - PubMed Central, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3084298/
  16. Updated literature review of agalsidase alfa (Replagal®) and agalsidase beta (Fabrazyme®) for the treatment of Fabry disease - PBS, accessed August 7, 2025, https://m.pbs.gov.au/info/industry/listing/elements/pbac-meetings/psd/2009-11/pbac-psd--agalsidase-alfa-literature-review.html
  17. Pegunigalsidase alfa: a novel, pegylated recombinant alpha-galactosidase enzyme for the treatment of Fabry disease - PMC, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11045972/
  18. Replagal, INN-agalsidase alfa - EMA, accessed August 7, 2025, https://www.ema.europa.eu/en/documents/product-information/replagal-epar-product-information_en.pdf
  19. Australian Public Assessment Report for Agalsidase alfa ghu - Therapeutic Goods Administration (TGA), accessed August 7, 2025, https://www.tga.gov.au/sites/default/files/auspar-replagal.pdf
  20. Agalsidase alfa: a review of its use in the management of Fabry disease - PubMed, accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/22946754/
  21. Agalsidase Alfa A Review of its Use in the Management of Fabry Disease - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/230795833_Agalsidase_Alfa_A_Review_of_its_Use_in_the_Management_of_Fabry_Disease
  22. go.drugbank.com, accessed August 7, 2025, https://go.drugbank.com/drugs/DB15874#:~:text=%CE%B1%2Dgalactosidase%20A%20is%20uptaken,the%20mannose%206%20phosphate%20receptor.&text=Agalsidase%20alfa%20hydrolyzes%20globotriaosylceramide%20and,by%20endogenous%20%CE%B1%2Dgalactosidase%20A.
  23. biochemical and pharmacological comparison of enzyme replacement therapies for the glycolipid storage disorder Fabry disease - Oxford Academic, accessed August 7, 2025, https://academic.oup.com/glycob/article/13/4/305/565768
  24. A biochemical and pharmacological comparison of enzyme replacement therapies for the glycolipid storage disorder Fabry disease - Oxford Academic, accessed August 7, 2025, https://academic.oup.com/glycob/article-pdf/13/4/305/1512074/cwg034.pdf
  25. annex i summary of product characteristics - Fabrazyme, INN-agalsidase beta, accessed August 7, 2025, https://ec.europa.eu/health/documents/community-register/2023/20230613159285/anx_159285_en.pdf
  26. Replagal | European Medicines Agency (EMA), accessed August 7, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/replagal
  27. Treatment of Fabry Disease: Established and Emerging Therapies ..., accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9967779/
  28. Pegunigalsidase alfa: a novel, pegylated recombinant alpha-galactosidase enzyme for the treatment of Fabry disease - Frontiers, accessed August 7, 2025, https://www.frontiersin.org/journals/genetics/articles/10.3389/fgene.2024.1395287/full
  29. An open-label clinical trial of agalsidase alfa enzyme replacement therapy in children with Fabry disease who are naïve to enzyme replacement therapy - PubMed, accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/27307708/
  30. Enzyme replacement therapy with agalsidase alfa in patients with Fabry's disease: an analysis of registry data - PubMed, accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/19959221/
  31. Safety and effectiveness of enzyme replacement therapy with agalsidase alfa in patients with Fabry disease: Post-marketing surveillance in Japan - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/331229175_Safety_and_effectiveness_of_enzyme_replacement_therapy_with_agalsidase_alfa_in_patients_with_Fabry_disease_Post-marketing_surveillance_in_Japan
  32. Long-term safety of enzyme replacement therapy with agalsidase alfa in patients with Fabry disease: post-marketing extension surveillance in Japan - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/382678151_Long-term_safety_of_enzyme_replacement_therapy_with_agalsidase_alfa_in_patients_with_Fabry_disease_post-marketing_extension_surveillance_in_Japan
  33. Agalsidase Alfa: Side Effects, Uses, Dosage, Interactions, Warnings, accessed August 7, 2025, https://www.rxlist.com/agalsidase_alfa/generic-drug.htm
  34. (PDF) The Safety of Agalsidase Alfa Enzyme Replacement Therapy in Canadian Patients with Fabry Disease Following Implementation of a Bioreactor Process - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/354721879_The_Safety_of_Agalsidase_Alfa_Enzyme_Replacement_Therapy_in_Canadian_Patients_with_Fabry_Disease_Following_Implementation_of_a_Bioreactor_Process
  35. Subject: Agalsidase Beta (Fabrazyme®) IV - Medical Coverage Guideline - Florida Blue, accessed August 7, 2025, https://mcgs.bcbsfl.com/MCG?mcgId=09-J2000-59&pv=false
  36. Agalsidase alfa versus agalsidase beta for the treatment of Fabry disease: an international cohort study - PMC, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5931248/
  37. Treatment of Fabry Disease: Outcome of a Comparative Trial with Agalsidase Alfa or Beta at a Dose of 0.2 mg/kg | PLOS One, accessed August 7, 2025, https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0000598
  38. Agalsidase alfa versus agalsidase beta for the treatment of Fabry disease: an international cohort study - PubMed, accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/29437868/
  39. Agalsidase alfa versus agalsidase beta for the treatment of Fabry disease: an international cohort study - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/322997021_Agalsidase_alfa_versus_agalsidase_beta_for_the_treatment_of_Fabry_disease_an_international_cohort_study
  40. Agalsidase alfa versus agalsidase beta for the treatment of Fabry disease: an international cohort study - UCL Discovery, accessed August 7, 2025, https://discovery.ucl.ac.uk/10044797/8/Elliott%20VoR351.full.pdf
  41. Replagal (agalsidase alfa): What is it and is it FDA approved ..., accessed August 7, 2025, https://www.drugs.com/history/replagal.html
  42. Agalsidase Alfa: Pediatric Medication | Memorial Sloan Kettering Cancer Center, accessed August 7, 2025, https://www.mskcc.org/cancer-care/patient-education/medications/pediatric/agalsidase-alfa
  43. Agalsidase Alfa - Drugs and Lactation Database (LactMed®) - NCBI Bookshelf, accessed August 7, 2025, https://www.ncbi.nlm.nih.gov/books/NBK535604/
  44. Agalsidase beta: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 7, 2025, https://go.drugbank.com/drugs/DB00103
  45. Shire application withdrawal disappoints Fabry disease foundation - MedCity News, accessed August 7, 2025, https://medcitynews.com/2012/03/shires-application-withdrawal-disappoints-fabry-disease-foundation/
  46. Shire withdraws Replagal in USA as FDA wants more trials - PharmaTimes, accessed August 7, 2025, https://pharmatimes.com/news/shire_withdraws_replagal_in_usa_as_fda_wants_more_trials_975565/
  47. www.sec.gov, accessed August 7, 2025, https://www.sec.gov/Archives/edgar/data/936402/000095010312001343/dp29324_ex9901.htm
  48. Shire withdraws application for FDA approval of Replagal - FirstWord Pharma, accessed August 7, 2025, https://firstwordpharma.com/story/1395865
  49. Shire Submits Biologics License Application (BLA) for REPLAGAL with the U.S. Food and Drug Administration (FDA) | Fierce Biotech, accessed August 7, 2025, https://www.fiercebiotech.com/biotech/shire-submits-biologics-license-application-bla-for-replagal-u-s-food-and-drug
  50. Agalsidase alfa for Fabry disease, accessed August 7, 2025, https://fabrydiseasenews.com/replagal/
  51. CENTER FOR DRUG EVALUATION AND RESEARCH - accessdata.fda.gov, accessed August 7, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2023/103979Orig1s5309.pdf
  52. Chiesi Global Rare Diseases and Protalix BioTherapeutics Announce Validation of Variation Submission by European Medicines Agency for pegunigalsidase alfa, accessed August 7, 2025, https://protalixbiotherapeutics.gcs-web.com/news-releases/news-release-details/chiesi-global-rare-diseases-and-protalix-biotherapeutics-2
  53. head trial of pegunigalsidase alfa versus agalsidase beta in patients ..., accessed August 7, 2025, https://jmg.bmj.com/content/61/6/534.full.pdf
  54. Head-to-head trial of pegunigalsidase alfa versus agalsidase beta in patients with Fabry disease and deteriorating renal function: results from the 2-year randomised phase III BALANCE study. - Apollo, accessed August 7, 2025, https://www.repository.cam.ac.uk/items/d05ea251-eaeb-430e-b131-98aa5000318c
  55. Head-to-head trial of pegunigalsidase alfa versus agalsidase beta in patients with Fabry disease and deteriorating renal function: results from the 2-year randomised phase III BALANCE study - PMC - PubMed Central, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11137442/
  56. (PDF) Response to commentary: Head-to-head trial of pegunigalsidase alfa versus agalsidase beta in patients with Fabry disease and deteriorating renal function: results from the 2-year randomised phase III BALANCE study – determination of immunogenicity - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/379676607_Response_to_commentary_Head-to-head_trial_of_pegunigalsidase_alfa_versus_agalsidase_beta_in_patients_with_Fabry_disease_and_deteriorating_renal_function_results_from_the_2-year_randomised_phase_III_BA
  57. Publication of Results from Phase 3 BALANCE Study in Fabry Disease, accessed August 7, 2025, https://chiesirarediseases.com/media/20240110-publication-of-results-from-phase-3-balance-study-in-fabry-disease
  58. Head-to-head trial of pegunigalsidase alfa versus agalsidase beta in patients with Fabry disease and deteriorating renal function: results from the 2-year randomised phase III BALANCE study | Journal of Medical Genetics, accessed August 7, 2025, https://jmg.bmj.com/content/61/6/520
  59. Agalsidase alfa Completed Phase 3 Trials for Fabry´s Disease Treatment | DrugBank Online, accessed August 7, 2025, https://go.drugbank.com/drugs/DB15874/clinical_trials?conditions=DBCOND0050158&phase=3&purpose=treatment&status=completed
  60. Agalsidase alfa Completed Phase 1 Trials for Fabry´s Disease Treatment | DrugBank Online, accessed August 7, 2025, https://go.drugbank.com/drugs/DB15874/clinical_trials?conditions=DBCOND0050158&phase=1&purpose=treatment&status=completed
  61. Substrate reduction therapy for Fabry disease, accessed August 7, 2025, https://fabrydiseasenews.com/substrate-reduction-therapy/
  62. Fabry Disease: Current and Novel Therapeutic Strategies. A Narrative Review - PubMed, accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/35652398/
  63. Novel treatment shows promise in cell model of Fabry disease | Substrate reduction therapy may help reduce Gb3S enzyme levels, accessed August 7, 2025, https://fabrydiseasenews.com/news/novel-srt-treatment-shows-promise-fabry-disease-cell-model/
  64. Fabry Disease and Gene Therapy - YouTube, accessed August 7, 2025, https://www.youtube.com/watch?v=UqPP732iJ-8

Published at: August 7, 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.