Next Article in Journal
The Implications of Type 1 Diabetes Mellitus Associated with Coeliac Disease
Previous Article in Journal
Reverse Total Shoulder Arthroplasty for Proximal Humerus Nonunion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Clinical Efficacy and Real-World Effectiveness of Fabry Disease Treatments: A Systematic Literature Review

1
The Mark Holland Metabolic Unit, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
2
Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
3
Global Evidence and Outcomes, Data and Quantitative Sciences Institute, Takeda Development Center Americas, Inc., Cambridge, MA 02142, USA
4
Oxford PharmaGenesis Ltd., Tubney OX13 5QJ, UK
5
Lysosomal Storage Disorders Unit, Royal Free London NHS Foundation Trust, University College London, London WC1E 6BT, UK
6
Vall d’Hebron Institute of Research, Vall d’Hebron Barcelona Hospital Campus, MPS-Lisosomales Medical Committee, 08035 Barcelona, Spain
7
Department of Genetics, Federal University of Rio Grande do Sul (UFRGS), Medical Genetics Service, Hospital de Clinicas de Porto Alegre (HCPA), Dasa Genomics, and Casa dos Raros, Porto Alegre 90610-261, Brazil
8
Nephrology Department, University Campus Bio-Medico, University of Rome, 00128 Rome, Italy
9
College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(14), 5131; https://doi.org/10.3390/jcm14145131
Submission received: 23 May 2025 / Revised: 2 July 2025 / Accepted: 9 July 2025 / Published: 18 July 2025
(This article belongs to the Section Endocrinology & Metabolism)

Abstract

Objectives: This systematic literature review aimed to identify studies assessing the clinical efficacy and real-world effectiveness of current and emerging treatments for Fabry disease. Methods: Searches of the MEDLINE, EMBASE, and Cochrane library databases, as well as relevant congress proceedings, were conducted to identify publications reporting on studies in patients of any age, sex, race, or ethnicity who received any approved or experimental treatment for Fabry disease, published before 17 June 2024. Results: Of 1881 publications screened, 234 reported data on renal, cardiac, cerebrovascular, and disease severity outcomes from 225 studies. The majority of reported studies were observational in nature (n = 150; 67%) and involved only adults (n = 172; 74%). Study designs and patient populations were highly heterogeneous, and cross-study conclusions about the effectiveness of different therapies could not be made. Enzyme replacement therapy (ERT) with agalsidase alfa or agalsidase beta stabilized renal function and cardiac structure in patients with Fabry disease. Early initiation of ERT in childhood or young adulthood was associated with better renal and cardiac outcomes than treatment initiation at a later age. The small number of comparator studies of agalsidase alfa and agalsidase beta suggested similar efficacy. Patients treated with migalastat and pegunigalsidase alfa also maintained stable renal function and cardiac structure. Conclusions: Overall, current treatments slow the progression of renal and cardiac decline in patients with Fabry disease. Large cohort studies with long-term follow-up and baseline stratification based on clinical phenotype are needed to address evidence gaps and provide clinicians with robust data to inform treatment decisions.

1. Introduction

Fabry disease (FD) is a rare, X-linked lysosomal storage disease caused by variants in the α-galactosidase A gene (GLA) that result in deficient α-galactosidase A enzyme (α-Gal A) activity [1]. This deficiency results in the systemic accumulation of glycosphingolipids in lysosomes and leads to heterogeneous, progressive manifestations, including renal failure, cardiomyopathy, and cerebrovascular disease, together with a significant reduction in life expectancy [1,2,3,4]. The severe ‘classic’ FD phenotype presents with multisystemic involvement, whereas ‘non-classic’ or ‘late-onset’ FD phenotypes may be restricted to a single organ, usually the heart [5,6]. The rate of disease progression can vary substantially among patients [7].
Treatment for FD involves the replacement or augmentation of α-Gal A function. Several approved therapies are available, including enzyme replacement therapy (ERT) with agalsidase alfa [8], agalsidase beta [9,10], or more recently, pegunigalsidase alfa (a pegylated form of the α-Gal A enzyme) [11,12], and oral chaperone therapy with migalastat [13,14] for patients with amenable GLA variants. Beyond these therapies, research into the treatment of FD continues to advance, with several emerging therapies under investigation in clinical trials. These include gene therapy to address the genetic defect responsible for the α-Gal A deficiency, and substrate reduction therapy, a novel approach intended to modulate the metabolic pathways involved in glycosphingolipid synthesis [15,16].
The purpose of this systematic literature review (SLR) was to identify and summarize the literature relating to the clinical efficacy (based on clinical trial data) and real-world effectiveness (based on data from observational studies) of available and emerging treatments for FD. Specific topics of interest were the impact of treatment on cardiovascular, renal, cerebrovascular, and disease severity outcomes.

2. Methods

2.1. Search Strategy

The SLR search strategy and reporting was fully compliant with the 2020 Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines [17]. The protocol was prospectively registered in the International Prospective Register of Systematic Reviews in Health and Social Care (PROSPERO, ID number CRD42023394203) on 26 January 2023. Searches of MEDLINE (in-process and other non-indexed citations), EMBASE, and the Cochrane library databases were conducted on 28 November 2022 and repeated using the same criteria on 17 June 2024. Search strings are shown in Supplementary Table S1. The population of interest was patients with FD of any age, sex, race, or ethnicity who received any approved or experimental disease-specific treatment for FD. Searches were restricted to English but were not limited by year or region. Case reports were excluded. Proceedings from the following key congresses (from 2020 to 2024) were also searched for relevant abstracts and posters: the Lysosomal Diseases Gordon Research Conference (GRC), the Society for the Study of Inborn Errors of Metabolism (SSIEM) annual symposium, the We’re Organizing Research on Lysosomal Diseases (WORLD) symposium, the International Congress of Inborn Errors of Metabolism (ICIEM), and the International Society for Pharmacoeconomics and Outcomes Research (ISPOR) annual conference.

2.2. Data Collection, Extraction, and Bias Assessment

Relevant abstracts and titles were identified by a single reviewer based on the criteria outlined in Supplementary Table S2. Articles meeting the inclusion criteria, or for which eligibility was unclear, were revisited for a full-text review by another single reviewer to confirm eligibility. Systematic reviews were included for full-text review to identify potentially relevant studies in their reference lists. Remaining uncertainties regarding eligibility were addressed by a separate reviewer. Relevant data from eligible publications were extracted to populate predefined summary tables, with all data being checked by a second reviewer. Extracted data included the study design, country, number of patients, baseline demographics, length of follow-up, treatment details, outcomes of interest (see below), funding information, and author conclusions. Publications were classified and summarized using the following categories: agalsidase alfa single-arm studies, agalsidase beta single-arm studies, mixed or non-specified ERT or other treatment single-arm studies, comparator studies, and studies examining the effects of switching between treatments (switch studies). For each included study, a risk of bias assessment was performed using the RoB2 tool for randomized trials, the Risk Of Bias In Non-randomized Studies—of Interventions (ROBINS-I) tool for non-randomized studies, or the Joanna Biggs Institute (JBI) critical appraisal checklist for systematic reviews and research synthesis (for meta-analyses).

2.3. Outcome Measures

The SLR strategy aimed to identify literature relating to the clinical efficacy and real-world effectiveness of different treatments for FD. Outcomes of interest were renal outcomes (glomerular filtration rate [GFR] and proteinuria); cardiovascular outcomes (left ventricular mass index [LVMI]/left ventricular mass [LVM] and other cardiac structure outcomes); cerebrovascular outcomes (white matter lesions [WMLs], stroke, transient ischemic attack [TIA], and other cerebrovascular outcomes); and the Mainz Severity Score Index (MSSI), a scoring system developed to measure the severity of general, neurological, cardiovascular, and renal signs and symptoms of FD. The SLR also identified safety outcomes data (adverse events and serious adverse events); these have not been included here owing to the heterogeneous nature of safety reporting across the included clinical studies.

3. Results

A flow diagram summarizing the search and screening process is presented in Figure 1. Overall, 1881 articles were identified from the electronic databases. An additional 20 relevant publications were identified in congress searches. After removing duplicates and non-eligible studies, 234 publications remained, covering a total of 225 studies. Most studies were real-world, observational studies (n = 150; 67%); the remaining studies were clinical trials (n = 71; 32%) and meta-analyses (n = 2; 1%). Geographically, study locations were wide-ranging, with the largest number conducted in Germany (n = 25) or across multiple countries (n = 70; Supplementary Figure S1). Most studies included only adults (n = 172; 74%). In studies that reported clinical outcomes, the number of included patients ranged from six [18,19] to 2051 [20]. Seven studies included over 500 patients [20,21,22,23,24,25,26]. The pharmaceutical industry funded 122 (54%) studies (41 studies were not industry funded; funding was not reported in the remaining 62 studies identified).
Overall, 187 studies included ERT with agalsidase alfa and/or agalsidase beta, 30 studies included treatment with migalastat, and 11 studies included second-generation ERT with pegunigalsidase alfa. Of the publications on single-arm studies (i.e., those without a comparator arm), 37 investigated the effects of agalsidase alfa, and 25 examined the effects of agalsidase beta. In addition, 23 publications examined mixed or non-specified ERT and 21 examined other treatments (including second-generation ERT) in single-arm studies. Overall, 32 publications compared various treatment regimens with each other or with untreated or placebo-treated patients. Only three studies compared the effects of agalsidase alfa and agalsidase beta [27,28,29]. In total, 20 publications examined the effects of switching between different treatments; this excluded investigations in which the effects of the switch were not reported as outcomes [30,31,32,33,34,35,36,37]. A summary of the risk of bias assessments is shown in Supplementary Table S3. Studies reported in congress publications were more likely to have a high risk of bias or lack information than studies reported in journal articles.

3.1. Renal Outcomes

FD causes glycosphingolipid accumulation and damage throughout the glomerular, vascular, and interstitial compartments of the kidney, resulting in progressive decline in kidney function and increased proteinuria [38]. Publications reporting GFR outcomes (reported as GFR, estimated GFR [eGFR], or measured GFR) presented data from single-arm studies of agalsidase alfa (n = 29) [26,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66], agalsidase beta (n = 19) [23,30,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83], mixed or non-specified ERT (n = 13) [31,35,36,84,85,86,87,88,89,90,91,92,93], other treatments (n = 18) [94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111], comparator studies (n = 21) [27,28,29,33,108,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127], and switch studies (n = 13) [128,129,130,131,132,133,134,135,136,137,138,139,140]. An overview of the GFR outcome data from agalsidase alfa and agalsidase beta single-arm studies and the comparator studies is presented in Table 1. Key results from the other types of studies are provided in Supplementary Table S4.
Treatment duration in the identified agalsidase alfa single-arm studies ranged from 26 weeks [51] to 20 years [40] (Table 1A). In 16 studies with follow-up durations of less than 5 years, treatment with agalsidase alfa demonstrated a renoprotective effect, stabilizing renal function (i.e., no statistically significant change in eGFR over the follow-up period) [39,44,46,48,49,50,51,54,56,58,59,60,61,62,63,64,65,66]. In 10 longer-term studies (>5 years), mixed results were reported [26,40,42,43,45,47,52,53,55,57]. In a retrospective, observational study, eGFR values remained stable over 10 years of treatment [47]. In a 10-year, prospective clinical study, weekly infusions of agalsidase alfa at a dose of 0.2 mg/kg were found to be beneficial for patients whose kidney function had declined with standard every-other-week dosing [53,57]. However, this study included small numbers of patients (n = 12). In two larger Fabry Outcome Survey (FOS) studies (n > 150), with longer follow-up periods (median treatment duration ≥ 12 years), agalsidase alfa treatment stabilized eGFR in patients with preserved kidney function (eGFR ≥ 60 mL/min/1.73 m2) or low proteinuria (≤0.5 g/24 h) at baseline [42,43]. In contrast, patients with impaired renal function (eGFR < 60 mL/min/1.73 m2) or high proteinuria (>0.5 g/24 h) at baseline continued to show declines in eGFR during treatment, although these declines were less than expected based on FD natural history data [42,43]. In a large, retrospective cohort study (n = 560) with a mean treatment duration of 7.6 years, patients who initiated agalsidase alfa treatment after 18 and 30 years of age had significant annual declines in mean (95% confidence interval [CI]) eGFR over the follow-up period (−1.12 [−1.77, −0.47] and −2.60 [−3.16, −2.04] mL/min/1.73 m2, respectively), whereas those who initiated treatment in childhood (≤18 years old) did not have significant annual changes in mean eGFR (−0.44 [−0.42, 1.30] mL/min/1.73 m2) [26]. In the longest agalsidase alfa study reported (in a congress publication), which included 66 patients from the FOS registry who had received agalsidase alfa for at least 19 years, the mean (standard deviation [SD]) annual rate of eGFR decline was −2.04 (0.36) mL/min/1.73 m2 (n = 48) [40].
Treatment duration in single-arm studies of agalsidase beta reporting GFR outcomes ranged from 22.7 months [68] to 10 years [82] (Table 1A). In total, 17of 19 identified studies reported a stabilizing effect of agalsidase beta on kidney function [23,30,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83], which varied based on factors, such as baseline GFR (based on clearance of 93Technetium DTPA [68] and eGFR [72]) and level of renal involvement [23,70,82]. In a long-term analysis (median treatment duration: 10 years) of patients with classic FD who participated in a phase 3 clinical trial of agalsidase beta, its extension study, and the Fabry Registry (n = 52), patients with low renal involvement at baseline (urine protein-to-creatinine ratio [UPCR] ≤ 0.5 g/g and <50% sclerotic glomeruli on kidney biopsy) had a mean annual eGFR decline of −1.89 mL/min/1.73 m2 [82]. In contrast, those with high renal involvement (UPCR > 0.5 g/g or ≥50% sclerotic glomeruli on kidney biopsy) had a mean annual eGFR decline of −6.82 mL/min/1.73 m2 despite treatment [82]. In patients from the Fabry Registry who initiated agalsidase beta treatment at a young age (5–30 years old; median follow-up: ≥5 years), annual eGFR decline was modest for both males (−1.18 mL/min/1.73 m2; n = 117) and females (−0.92 mL/min/1.73 m2; n = 59) with low renal involvement (UPCR ≤ 0.5 g/g or urine albumin-to-creatinine ratio [UACR] ≤ 0.3 g/g) at baseline, whereas males with high renal involvement (UPCR > 0.5 g/g or UACR > 0.3 g/g) showed a greater annual decline in eGFR (−2.39 mL/min/1.73 m2; n = 23) [23].
Single-arm studies evaluating mixed or non-specified ERT also demonstrated the importance of early initiation of ERT for better long-term renal outcomes (Supplementary Table S4). In a retrospective cohort of adults receiving ERT (n = 293; median follow-up: 6.8 years), those with lower eGFR at baseline had an increased risk of having a first renal event (hazard ratio [per −10 mL/min/1.73 m2]: 5.57, p = 0.02) [31]. In single-arm studies of other treatments, 12 studies (13 publications) specifically evaluated the effects of migalastat [14,97,98,99,100,101,103,104,105,106,108,109,110], four examined second-generation ERT with pegunigalsidase alfa [94,95,96,107], and one explored the effects of a lentivirus-mediated gene therapy [102] on eGFR in patients with FD. Renal function remained stable in most migalastat studies (follow-up periods ranged from 6 months to 7 years) [98,99,100,101,105,106,108,110,111]. In a prospective, observational study of migalastat treatment over 24 months (n = 59), mean (SD) eGFR significantly decreased from 98.6 (15.2) to 90.4 (14.7) mL/min/1.73 m2 (p = 0.0317) in females and from 99.8 (22.6) to 92.4 (24.0) mL/min/1.73 m2 (p = 0.0028) in males [103,104]. In the four studies that examined the effects of pegunigalsidase alfa (n = 15–30), eGFR remained stable over follow-up periods of 3 to 72 months [94,95,96,107]. In the single study of lentivirus-mediated gene therapy, five patients were infused with autologous lentivirus-transduced, CD34+-selected, hematopoietic stem/progenitor cells engineered to express α-Gal A [102]. Patients had a transient increase in eGFR during the active treatment phase, although eGFR returned to baseline levels and remained stable after gene therapy (follow up: 12–33 months), except for one patient who displayed progressive chronic kidney disease with significant proteinuria at baseline [102].
In total, 12 studies reporting on GFR outcomes compared active treatments with no treatment or placebo [33,108,112,114,115,116,117,120,121,122,123,124,126], 10 studies directly compared different active treatments [27,28,29,108,113,114,115,119,122,125,127] (two studies included active comparator groups and untreated groups [112,114]), and two compared different doses of the same treatment [33,118] (Table 1B). Overall, studies that included patients who received no treatment or placebo indicated that approved treatments for FD generally resulted in stabilization of GFR outcomes, although study designs, patient populations, and follow-up times varied considerably. In a comparison of FOS registry data (n = 740; median treatment duration: 5.4 years) and published findings for untreated patients with FD, treatment with agalsidase alfa slowed eGFR decline in male patients with low eGFR (<60 mL/min/1.73 m2) at baseline [112]. A stabilizing effect of ERT on eGFR was also observed in patients with FD following kidney transplantation [113,114,115]. Three studies (two cohort studies and one randomized controlled open-label trial) directly compared agalsidase alfa and agalsidase beta with respect to effects on eGFR; no significant differences between the two treatments were identified [27,28,29]. In the largest of these studies, there was no significant difference in the slope of eGFR between patients treated with agalsidase alfa (n = 248) or agalsidase beta (n = 139) over a median follow-up period of 4.9 years. This finding was seen regardless of baseline eGFR (≥60 or <60 mL/min/1.73 m2) and after adjusting for additional factors including sex, phenotype, and baseline proteinuria [29]. In the phase 3 BALANCE trial, pegunigalsidase alfa showed noninferiority to agalsidase beta based on the rate of eGFR decline over 24 months in adults with deteriorating renal function at baseline (linear eGFR slope more negative than −2 mL/min/1.73 m2/year; n = 52) [127].
Ten studies (17 publications) assessed the effects of switching from one FD treatment to another on GFR outcomes (Supplementary Table S4), of which six studies (seven publications) focused on patients switching from agalsidase beta to agalsidase alfa following an agalsidase beta supply shortage from June 2009 to January 2012 [128,131,132,133,136,137,138]. Four studies showed mild deterioration in eGFR after switching from the standard dosing of agalsidase beta to agalsidase alfa (n = 22–38) [131,132,133,138]. However, all of these studies were observational and nonrandomized; therefore, treatment groups were not homogeneous. Two other studies demonstrated maintenance of eGFR in patients following the switch from agalsidase beta to agalsidase alfa (n = 71 and 11) [128,136,137]. In the phase 3 BRIDGE trial (n = 20), pegunigalsidase alfa slowed progression of kidney disease (mean eGFR slope was −5.90 mL/min/1.73 m2/year with agalsidase alfa compared with −1.19 mL/min/1.73 m2/year 12 months after switching to pegunigalsidase alfa) [139]. The small number of patients means the data could be influenced by a few patients. There was no difference in GFR outcomes in studies in which patients were switched from ERT to migalastat [129,130,134,135,140].
Identified studies reporting on proteinuria in patients receiving FD treatments (42 studies in 44 publications) are shown in Supplementary Table S5. Overall, the majority of studies showed little to no effect of treatment on proteinuria in patients with FD, although improvements in at least one proteinuria outcome were reported in four out of the six studies in children [33,51,76,81]. However, supportive treatment with an angiotensin-converting enzyme inhibitor (ACEi) or angiotensin receptor blocker (ARB) is standard practice for any patient with proteinuric kidney disease, and has been shown to provide sustained reductions in proteinuria and stabilization of kidney function in a small study of patients with severe FD [79]. The differing use of supportive treatments with ACEi and ARB may have affected the results from other studies and cannot be independently assessed. The additional effect of sodium-glucose co-transporter-2 inhibitors combined with ERT on proteinuria and renal outcomes in FD remains to be established.
Table 1. Overview of GFR data from (A) single-arm agalsidase alfa or agalsidase beta studies and (B) comparator studies.
Table 1. Overview of GFR data from (A) single-arm agalsidase alfa or agalsidase beta studies and (B) comparator studies.
(A)
Author Year
Study Identifier
NTreatment DurationKey Results
Agalsidase alfa single-arm studies
Beck 2004 [39]
FOS
150Mean [max]:
17 [56] months
  • Observational study using data from FOS
  • In patients with a baseline GFR between 60 and 90 mL/min/1.73 m2 (n = 104), GFR was stable following 1 year (n = 57) and 2 years (n = 30) of treatment
  • Patients with a baseline GFR between 30 and 60 mL/min/1.73 m2 (n = 46) also had stable GFR at 1 year (n = 26) and 2 years (n = 18) of treatment
Cybulla 2022 [43]
FOS
193Mean/median (SD) [range]: 12.6/11.9 (5.0) [5.0, 21.6] years
  • Observational study using male patient data from FOS
  • eGFR (mL/min/1.73 m2)
    Low proteinuria (n = 114): intercept (SE), 9.67 (4.00); slope (SE), −1.61 (0.28); p < 0.0001
    High proteinuria (n = 51): intercept (SE), 4.15 (3.69); slope (SE), −3.62 (0.42); p < 0.0001
    p (high vs. low proteinuria) < 0.0001
Feriozzi 2009 [44]
FOS
1653 years
  • Observational study using data from FOS
  • eGFR remained stable in men with stage III disease; in those with stage I or II renal disease, there was a small but significant decrease in eGFR (p < 0.01)
  • The mean decrease in eGFR in all male patients (n = 115) was 2.66 ± 5.07 mL/min/1.73 m2/year
  • The decline in eGFR (SD) was numerically larger in patients with urine protein levels > 500 mg/24 h (n = 14) −3.98 (7.86) mL/min/1.732/year than in patients with urine protein levels <500 mg/24 h (n = 40) −1.68 (3.59) mL/min/1.732/year
Feriozzi 2012 [45]
FOS
208Mean [range]:
7.4 [5.0, 11.2] years
  • Observational study using data from FOS
  • Mean (95% CI) yearly change in eGFR was −2.2 (−2.8, −1.7) mL/min/1.73 m2 in men (n = 134) and −0.7 (−1.4, 0.0) mL/min/1.73 m2 in women (n = 74)
Giugliani 2023 * [40]
FOS
66Median [IQR]: 20.03 [19.55, 20.64] years
  • Observational study using data from FOS
  • Baseline eGFR: 99.35 (27.07) mL/min/1.73 m2
  • Annual rate of change: mean (SE), −2.04 (0.36) mL/min/1.73 m2 (n = 48)
  • Mean reduction: males (n = 38), −2.27 mL/min/1.73 m2; females (n = 10), −1.24 mL/min/1.73 m2
  • Statistical significance values not reported
Goker-Alpan 2016 [46]14Median [range]: 54.5 [54.0, 59.0] weeks
  • An open-label trial of agalsidase alfa in children
  • Mean (95% CI) change from baseline at week 55 of 0.15 (−11.39, 11.70) mL/min/1.73 m2 (median, 0.72 [−27.5, 35.9] mL/min/1.73 m2)
Hughes 2011 [48]
FOS
250≥4 years
  • Observational study using data from FOS
  • Median [10–90th percentile] eGFR (MDRD) at baseline (mL/min/1.73 m2): women, 71.8 [56.5–87.6]; men, 88.2 [41.1–137.0]
  • Median [10–90th percentile] eGFR (MDRD) at 4 years (mL/min/1.73 m2): women (n = 41), 69.6 [44.8–91.1], p = 0.007 vs. baseline;
    men (n = 105), 80.3 [34.2–119.7], p < 0.001 vs. baseline
Kampmann 2015 [47]45Median [range]: 10.8 [9.6, 12.5] years
  • Single-center observational study in Germany
  • Among patients (8 males, 5 females) with pretreatment eGFR ≥ 90 mL/min/1.73 m2, the mean annual decline in eGFR over 10 years was not statistically significant
  • In patients with poorer renal function (10 males, 11 females), eGFR seemed to improve in the first 3 years of ERT
  • After 10 years, however, eGFR values were not significantly changed regardless of renal function before ERT
Kleinert 2006 [64]
FOS
60>2 years
  • Observational study using data from FOS
  • Median [10th–90th percentile] eGFR (mL/min/1.73 m2): baseline: 88 [49–126] (n = 54); 1 year: 86; 2 years: 84
Mehta 2009 [65]
FOS
181Mean (SD):
3.1 (2.1) years
  • Observational study using data from FOS
  • All patients: mean (SD) eGFR (mL/min/1.73 m2)
    Year 1 (n = 121); baseline: 85.46 (29.21); year-end: 82.31 (29.12); change: −3.15 (13.01); annualized: −3.15; p = 0.01
    Year 2 (n = 143); baseline: 86.17 (27.65); year-end: 79.03 (29.47); change: −7.14 (16.82); annualized: −3.57; p = 0.001
    Year 3 (n = 137); baseline: 87.51 (28.63); year-end: 76.77 (27.01); change: −10.74 (14.91); annualized: −3.58; p = 0.001
    Year 5 (n = 150); baseline: 87.04 (28.80); year-end: 74.75 (28.65); change: −12.29 (18.40); annualized: −2.46; p = 0.001
  • Men: mean (SD) eGFR (mL/min/1.73 m2)
    Year 1 (n = 80); baseline: 94.02 (30.56); year-end: 90.30 (31.02); change: −3.72 (14.84); annualized: −3.72; p = 0.028
    Year 2 (n = 99); baseline: 93.23 (28.37); year-end: 85.58 (31.60); change: −7.65 (19.05); annualized: −3.83; p = 0.0001
    Year 3 (n = 92); baseline: 96.26 (29.01); year-end: 82.65 (27.91); change: −13.61 (14.66); annualized: −4.54; p < 0.0001
    Year 5 (n = 103); baseline: 94.71 (29.77); year-end: 78.84 (31.08); change: −15.87 (20.04); annualized: −3.17; p < 0.0001
  • Women: mean (SD) eGFR (mL/min/1.73 m2)
    Year 1 (n = 41); baseline: 68.76 (16.81); year-end: 66.71 (16.33); change: −2.05 (8.45); annualized: −2.05; p = NS
    Year 2 (n = 44); baseline: 70.28 (17.81); year-end: 64.30 (16.46); change: −5.98 (10.27); annualized: −2.99; p = 0.0004
    Year 3 (n = 45); baseline: 69.61 (17.49); year-end: 64.74 (20.59); change: −4.87 (13.80); annualized: −1.62; p = 0.02
    Year 5 (n = 47); baseline: 70.23 (17.37); year-end: 65.80 (19.96); change: −4.44 (10.72); annualized: −0.89; p = 0.007
  • Hyperfiltration (≥130 mL/min/1.73 m2): mean (SD) eGFR (mL/min/1.73 m2)
    Year 1 (n = 11); baseline: 144.49 (13.40); year-end: 131.13 (21.61); change: −13.35; annualized: −13.35; p = 0.05
    Year 2 (n = 12); baseline: 139.64 (8.75); year-end: 118.04 (43.46); change: −21.60; annualized: −10.80; p = NS
    Year 3 (n = 13); baseline: 142.47 (13.19); year-end: 114.36 (16.56); change: −28.11; annualized: −9.37; p = 0.001
    Year 5 (n = 14); baseline: 143.50 (13.25); year-end: 108.07 (20.26); change: −35.43; annualized: −7.09; p = 0.001
  • CKD Stage 1 (90–129 mL/min/1.73 m2): mean (SD) eGFR (mL/min/1.73 m2)
    Year 1 (n = 35); baseline: 107.06 (11.15); year-end: 103.94 (18.37); change: −3.12; annualized: −3.12; p = NS
    Year 2 (n = 47); baseline: 106.89 (10.34); year-end: 99.06 (19.38); change: −7.83; annualized: −3.91; p = 0.01
    Year 3 (n = 45); baseline: 107.63 (10.34); year-end: 95.37 (19.49); change: −12.26; annualized: −4.09; p = 0.001
    Year 5 (n = 48); baseline: 107.06 (10.30); year-end: 93.93 (23.48); change: −13.13; annualized: −2.63; p = 0.001
  • CKD Stage 2 (60–89 mL/min/1.73 m2): mean (SD) eGFR (mL/min/1.73 m2)
    Year 1 (n = 55); baseline: 73.61 (8.59); year-end: 72.21 (12.32); change: −1.39; annualized: −1.39; p = NS
    Year 2 (n = 60); baseline: 74.16 (8.73); year-end: 69.33 (14.03); change: −4.83; annualized: −2.41; p = 0.001
    Year 3 (n = 58); baseline: 73.25 (8.54); year-end: 66.84 (13.71); change: −6.40; annualized: −2.13; p = 0.001
    Year 5 (n = 64); baseline: 73.98 (8.74); year-end: 66.33 (17.26); change: −7.64; annualized: −1.53; p = 0.001
  • CKD Stage 3 (30–59 mL/min/1.73 m2): mean (SD) eGFR (mL/min/1.73 m2)
    Year 1 (n = 19); baseline: 48.78 (8.23); year-end: 45.92 (9.88); change: −2.87; annualized: −2.87; p = 0.05
    Year 2 (n = 23); baseline: 49.76 (7.93); year-end: 45.23 (9.82); change: −4.54; annualized: −2.27; p = 0.01
    Year 3 (n = 21); baseline: 49.77 (8.31); year-end: 41.07 (10.19); change: −8.70; annualized: −2.90; p = 0.001
    Year 5 (n = 23); baseline: 49.76 (7.93); year-end: 39.53 (14.50); change: −10.23; annualized: −2.05; p = 0.001
  • CKD Stage 4 (15–29 mL/min/1.73 m2): mean (SD) eGFR (mL/min/1.73 m2)
    Year 1 (n = 1); baseline: 28.78; year-end: 34.60; change: +5.81; annualized: +5.81; p = NA
    Year 2 (n = 1); baseline: 28.78; year-end: 29.29; change: +0.50; annualized: +0.25; p = NA
    Year 3 (n = 0); data: NR
    Year 5 (n = 1); baseline: 28.78; year-end: 37.05; change: +8.27; annualized: +1.65; p = NA
Parini 2020 [26]
FOS
560Mean (SD):
7.6 (4.8) years
  • Observational study using data from FOS
  • Overall mean eGFR (mL/min/1.73 m2): cohort 1: ≤18 years, 117.9; cohort 2: 18–30 years, 114.1; cohort 3: >30 years, 79.9
  • Changes during follow-up
    Cohort 1: ≤ 18 years; no significant annual changes in eGFR slope (95% CI) −0.44 mL/min/1.73 m2 (−0.42, 1.30) proteinuria was observed throughout the follow-up period
    Cohort 2: 18–30 years; although proteinuria remained stable, eGFR showed a significant annual decline over the follow-up period: eGFR slope (95% CI), −1.12 mL/min/1.73 m2 (−1.77, −0.47; p < 0.001)
    Cohort 3: >30 years; a significant annual deterioration over the follow-up period was shown for eGFR (slope (95% CI), −2.60 mL/min/1.73 m2 (−3.16, −2.04; p < 0.001)
Pastores 2007 [49]22Median [range]: 42 [11, 65] weeks
  • A phase 2, multi-center, open-label study
  • Patients with FD and ESRD undergoing dialysis or who had a history of kidney transplantations were treated with agalsidase alfa
  • After 26 weeks of agalsidase alfa therapy, mean (SEM) eGFR increased slightly to 71.6 (8.0) mL/min/1.73 m2 (n = 10) from 64.5 (6.0) mL/min/1.73 m2 (n = 12), which approached statistical significance in this relatively small cohort of patients (p = 0.07)
Pintos-Morell 2023 [41]
FOS
285Mean (SD):
Early initiators group: 11.2 (5.7) years;
Late initiators group: 11.8 (5.8) years
  • Observational study using data from FOS
  • Rates of annual eGFR change; mean (SE)
    Early-initiators (<25 years at treatment start) group (n = 40): −1.45 (0.52) mL/min/1.73 m2/year; p = 0.0054
    Late-initiators (≥25 years at treatment start) group (n = 103): −2.84 (0.32) mL/min/1.73 m2/year; p < 0.0001
  • The rate of eGFR change was significantly slower in the early initiators compared with the late initiators group; p = 0.0234
Ramaswami 2011 [50]
FOS
8Mean (SD):
4.2 (1.9) years
  • Observational study in children using data from FOS
  • Study of patients in the FOS registry who received their first dose of agalsidase alfa before reaching the age of 7 years
  • Mean (SD) baseline eGFR (mL/min/1.73 m2): 124.4 (25.7); final eGFR, 112.7 (21.79)
Ramaswami 2012 [66]
FOS
98≥6 months
  • Observational study in pediatric patients (<18 years old) using data from FOS
  • Mean (SD) eGFR at baseline and 12 months (n = 40): 96.8 (26.5) and 94.2 (21.2) mL/min/1.73 m2
  • Mean (SD) eGFR at baseline and 24 months (n = 45): 93.2 (26.1) and 92.5 (25.2) mL/min/1.73 m2
Ramaswami 2019 [42]
FOS
152Median [range]:
Evaluable treated renal cohort
(n = 152) 13.3 [10.1, 18.4] years;
Females (n = 62) 12.5 [10.1, 17.1] years;
Males (n = 90): 14.4 [10.1, 18.4] years
  • Observational study in children using data from FOS
  • Pediatric patients who had received agalsidase alfa ERT over a 10-year period
  • Mean (SD) baseline eGFR (mL/min/1.73 m2): evaluable treated renal cohort, 89.04 (26.49)
  • Number of patients (%) in eGFR category <60 mL/min/1.73 m2: evaluable treated renal cohort, 21/152 (13.8%)
  • Mean [95% CI] eGFR at year 10 (mL/min/1.73 m2): females 79.1 [74.1, 84.0]; males 77.5 [70.1, 84.9]
  • Mean [95% CI] eGFR slope in patients in eGFR category ≥ 60 mL/min/1.73 m2 at baseline: females (n = 52), −0.55 [−1.12, 0.01]; males (n = 79), −1.99 [−2.45, −1.54]
  • Mean [95% CI] eGFR slope in patients in eGFR category <60 mL/min/1.73 m2 at baseline: females (n = 10), −0.14 [−1.43, 1.15]; males (n = 11), −2.79 [−4.01, −1.56]
Ries 2006 [51]2426 weeks
  • Open-label study in pediatric patients
  • Study in pediatric patients with FD (between 6.5 and 18 years of age) naive to ERT
  • Mean (SEM) baseline eGFR was 121 (5.0) mL/min/1.73 m2
  • 7 patients had eGFR > 135 mL/1.73 m2
  • 2 patients showed evidence of kidney dysfunction defined as eGFR between 60 and 89 mL/min/1.73 m2
  • Mean (SEM) eGFR did not change significantly during the treatment (mL/min/1.73 m2): 9 weeks, 119.2 (3.6); 17 weeks, 120.0 (3.9); and 26 weeks, 116.0 (3.9)
  • The eGFR of patients with possible hyperfiltration (eGFR > 135 mL/min/1.73 m2) returned to the reference range
Sasa 2019 [52]493Mean [range]:
3.5 [0.0, 7.9] years;
<0.5: (n = 35);
>0.5–1: (n = 31);
>1–2: (n = 48);
>2–3: (n = 69);
>3–4: (n = 86);
>4–5: (n = 132);
>5–6: (n = 55);
>6–7: (n = 20);
>7–8: (n = 17)
  • A post-marketing surveillance study in Japan
  • Mean (SD) yearly change in eGFR (mL/min/1.73 m2)
    Males (classic phenotype): year 0.5, 1.00 (p = 0.619); year 1, −1.03
    (p = 0.289); year 2, −1.79 (p = 0.002); year 3, −2.09 (p = 0.001); year 4, −2.04 (p = 0.000); year 5, −2.50 (p = 0.000); year 6, −2.88 (p = 0.124); year 7, −0.44 (p = 0.814)
    Males (non-typical variant): year 0.5, −1.17 (p = 0.718); year 1, −1.24
    (p = 0.544); year 2, −1.73 (p = 0.103); year 3, −2.04 (p = 0.028); year 4, −1.47 (p = 0.057); year 5, −0.95 (p = 0.447); year 6, −1.54 (p = 0.084); year 7, NR
    Females: year 0.5, −1.81 (p = 0.328); year 1, −1.61 (p = 0.059); year 2, −1.44 (p = 0.008); year 3, −1.02 (p = 0.020); year 4, −1.10 (p = 0.028); year 5, −1.89 (p = 0.009); year 6, −1.95 (p = 0.021); year 7, −2.64 (p = 0.078)
Schiffmann 2006 [54]25≤54 months
  • Open-label extension study for male patients
  • Assessment of long-term renal effects and practicality of home infusions of agalsidase alfa
  • Data reported for 24/25 patients: 12 had stage I renal disease (eGFR, ≥ 90 mL/min/1.73 m2) at baseline, 8 had stage II (eGFR, 60–89 mL/min/1.73 m2), and 4 had stage III (GFR, 30–59 mL/min/1.73 m2) renal disease
  • Overall population: after 48 months, eGFR declined to 75.1 (32.7) mL/min/1.73 m2, p = 0.039 vs. baseline, mostly owing to changes in patients with stage 3 CKD
Schiffmann 2007 [53], Schiffmann 2015 [57]12≤10 years
  • Open-label single-center study in the US
  • Long-term prospective clinical trial assessing the effects of EOW versus weekly agalsidase alfa infusions in patients who experienced ongoing decline in renal function of EOW dosing
  • Before therapy with agalsidase alfa, mean (SD) eGFR was 77.8 (30.4) mL/min/1.73 m2
  • During EOW dosing, the mean (SD) rate of change of eGFR was −8.0 (2.8) mL/min/1.73 m2/year (−0.67 (0.23) mL/min/1.73 m2/month)
  • During 2 years of weekly dosing (following EOW dosing), the mean rate of change of eGFR slowed to −3.3 ± 4.7 mL/min/1.73 m2/year (−0.27 ± 0.40 mL/min/1.73 m2/month); p = 0.01
  • After 10 years of follow-up, mean (SD) slope on agalsidase alfa every 2 weeks was −7.92 (2.88) and 3.84 (4.08) mL/min/1.73 m2/year on weekly enzyme infusions (p = 0.01)
Schiffmann 2013 [56]7312 months
  • Data pooled from three randomized, double-blind, placebo-controlled trials and their open-label extension studies
  • 25 patients had stage II/III renal disease (baseline eGFR 30–90 mL/min/1.73 m2
  • Mean (SD) eGFR at baseline was 92.6 (32.4) mL/min/1.73 m2 and remained stable at 92.1 (32.4) at 12 months
Schiffmann 2014 [55]17Mean (SD):
6.5 (0.6) years
  • Open-label multi-center extension study for pediatric patients
  • Evaluating safety and efficacy of long-term agalsidase alfa ERT in children with FD
  • Pediatric patients with FD (7–17 years of age at study enrollment) received agalsidase alfa
  • Mean (SD) baseline eGFR was 123.29 (16.00) mL/min/1.73 m2
  • Most individuals showed relatively stable eGFR over time
Schwarting 2006 [58]
FOS
202 years
  • Observational study using data from FOS
  • eGFR (mL/min/1.73 m2), median [10th–90th percentile]
    Baseline: CKD stage 2 (n = 12), 70 [63–83]; CKD stage 3 (n = 8), 47
    [35–55]
  • Renal function in 12 patients (10 females and 2 males; mean age: 40 years; range: 19, 71 years) with baseline CKD stage 2 declined significantly
    (p < 0.05) in the year before treatment began
  • Renal function in 8 patients (4 females and 4 males; mean age: 49 years; range: 29, 69 years) with CKD Stage 3 also declined in the year before the start of treatment, but this change was not significant
  • After 1 year of treatment, however, the progressive loss of GFR had been stabilized in both groups of patients
  • Renal function remained stable in 8 patients with CKD stage 2 and in 5 patients with CKD stage 3 studied for 2 years after the start of ERT
Schwarting 2006b [59]
FOS
401>2 years
(n = 219)
>3 years
(n = 133)
  • Observational study using data from FOS
  • Mean, median [range] eGFR (mL/min/1.73 m2)
    Patients given 1 year of therapy with agalsidase alfa
    -
    Female (n = 52): baseline: 66.8, 66.7 [32.0, 89.5]; year 1: 65.2, 64.5 [29.3, 87.4]
    -
    Male (n = 47): baseline: 66.0, 68.3 [38.4, 89.5]; year 1: 63.2, 61.7 [26.1, 113.0]
    Patients given 2 years of therapy with agalsidase alfa
    -
    Female (n = 34): baseline: 65.6, 64.3 [32.0, 89.5]; year 2: 63.1, 63.5 [30.0, 97.7]
    -
    Male (n = 28): baseline: 69.4, 73.1 [39.4, 89.5]; year 2: 63.3, 63.0 [12.8, 92.4]
    Patients given 3 years of therapy with agalsidase alfa
    -
    Female (n = 13): baseline: 68.4, 64.2 [47.6, 89.5]; year 3: 66.3, 65.6 [48.9, 88.2]
    -
    Male (n = 12): baseline: 65.6, 69.3 [38.4, 84.5]; year 3: 60.6, 62.4 [23.9, 91.2]
Thofehrn 2009 [60]936 months (n = 6)
18 months (n = 2)
12 months (n = 1)
  • Single-center observational study in Brazil
  • Mean (SD) [range] GFR (mL/min/1.73 m2)
    Baseline (n = 8): 110.30 (28.68) (1 patient progressed to ESRD and underwent transplant, excluded from analysis of renal function)
    12 months (n = 8): 114.78 (33.92)
    24 months (n = 5): 104.38 (35.66)
    36 months (n = 5): 122.61 (46.85)
  • No significant (p > 0.05) differences were found
Tsuboi 2017 [61]
Japan Fabry Research—002
36Median [range]: 62.5 [8, 84] months
  • Prospective observational study in Japan
  • Cohort included ERT treatment-naive patients and those who were receiving agalsidase alfa 0.2 mg/kg EOW
  • Mean (SD) baseline eGFR: 83.5 (28.6); after follow-up: 82.6 (29.4) mL/min/1.73 m2
West 2009 [62]108Mean, median (SD):
2.0, 1.6 (1.0) years
Mean (SD) treatment years by baseline GFR range
(mL/min/1.73 m2)
≥135
(n = 8): 1.2 (0.2);
90 to <135
(n = 33): 2.0 (1.0);
60 to <90
(n = 36): 2.2 (1.2);
30 to <60
(n = 14): 2.1 (0.9)
15 to <30
(n = 2): 1.0
  • Summary of 3 prospective, randomized, placebo-controlled trials and their open-label extension studies
  • Mean (SD) [median; range] GFR (mL/min/1.73 m2):
    Baseline before placebo: 88.9 (32.5) [92.3; 12.7, 160]
    After 6 months of placebo: 85.0 (37.6) [83.3; 12.5, 184]
    Mean (SD) annualized rate of change in GFR: −7.7 (38.0) mL/min/1.73 m2/year; p = 0.14
  • 3 patients had renal hyperfiltration (i.e., baseline GFR > 135 mL/min/1.73m2)
  • When these 3 patients were removed from the analysis of the placebo period:
    Mean (SD) GFR at baseline before placebo (mL/min/1.73 m2): 85.4 (29.6)
    Mean (SD) GFR after 6 months of placebo (mL/min/1.73 m2): 81.9 (34.8)
    Mean (SD) annualized rate of change in GFR −7.0 (32.9) mL/min/1.73 m2/year; p = 0.12
  • Mean (SD) [median; range] GFR at baseline before agalsidase alfa (mL/min/1.73 m2): 90.3 (31.2) [87.4; 25–184]
  • Mean (SD) baseline GFR by baseline GFR range (mL/min/1.73 m2): ≥135 (n = 8), 151.9 (16.4); 90 to <135 (n = 33), 109.6 (13.4); 60 to <90 (n = 36), 78.1 (8.4); 30 to <60 (n = 14), 50.1 (8.2); 15 to <30 (n = 2), 26.7
  • Mean (SD) annualized rate of change in GFR for the entire study population: −4.8 (10.6) mL/min/1.73 m2/year (p = 0.0003 compared with no change); median, −4.1 mL/min/1.73 m2/year
  • After removing 8 patients with hyperfiltration during agalsidase alfa therapy: rate of change in GFR, −2.9 (8.7) mL/min/1.73 m2/year (p = 0.0002 compared with no change)
Whybra 2009 [63]364 years
  • Prospective single-center, open-label study of female patients in Germany
  • A cohort of female patients with FD who were experiencing manifestations of FD and had not been previously treated with ERT
  • Mean (SD) eGFR after 4 years of treatment: 91.0 (25.6) mL/min/1.73 m2
    4 patients with eGFR > 135 mL/min/1.73 m2 at baseline (defined as hyperfiltration): eGFR at after 4 years of treatment, 128.3 (4.6) mL/min/1.73 m2
  • Mean (SD) eGFR by baseline eGFR range (mL/min/1.73 m2; * p < 0.05 vs. baseline; ** p < 0.001 vs. baseline):
    eGFR > 135 (n = 4): month 12, 140.3 (26.8); month 24, 135.3 (18.3) *; month 36, 133.0 (17.0) *; month 48, 128.3 (4.6)
    eGFR 90–135 (n = 9): month 12, 98.7 (15.6); month 24, 98.5 (10.9); month 36, 106.7 (15.5); month 48, 100.4 (16.4)
    eGFR 60–89 (n = 20): month 12, 85.5 (13.2); month 24, 88.8 (14.5); month 36, 83.0 (17.9); month 48, 85.9 (20.2)
    eGFR 30–59 (n = 3): month 12, 50.4 (10.8); month 24, 55.8 (8.1); month 36, 49.7 (11.5); month 48, 47.3 (14.0)
  • Responder definitions: I, less than 20% decrease in GFR from baseline; II, no shift to more severe CKD stage eGFR 90–135 mL/min/1.73 m2 at baseline (n = 9): responder definition I (%), 89; responder definition II (%), 89
    eGFR 60–89 mL/min/1.73 m2 at baseline (n = 20): responder definition I (%), 95; responder definition II (%), 95
    eGFR 30–59 mL/min/1.73 m2 at baseline (n = 3): responder definition I (%), 67; responder definition II (%), 100
  • Patients treated with ACEIs or ARBs during entire study (n = 7)
    Mean (SD): eGFR after 4 years of treatment, 65.0 (13.5) mL/min/1.73 m2; proteinuria after 4 years of treatment, 425 (531) mg/24 h
  • Patients who began treatment with ACEIs or ARBs after starting treatment with agalsidase alfa (n = 6)
    Mean (SD): eGFR at last measurement before starting anti-angiotensin therapy, 87.7 (29.6) mL/min/1.73 m2; eGFR at study end, 83.0 (33.4) mL/min/1.73 m2
Agalsidase beta single-arm studies
Benichou 2009 [67]76Median [range]:
Phase 3: 55.9
[3.3, 60.7] months;
Phase 4: 33.5
[1.9, 51.2] months
  • Retrospective analysis of efficacy data obtained from adult patients with FD who received agalsidase beta for up to 5 years during a phase 3 or phase 4 placebo-controlled trial and the associated open-label extension study
  • No statistically significant correlation was found between eGFR slope and peak titer (the highest anti-aGAL IgG titer observed for a given patient during treatment) among male patients from either the phase 3 (r2 = −0.22, n = 56, p = 0.10) or phase 4 studies (r2 = −0.09, n = 66, p = 0.46)
  • Similar correlation coefficients were found when both males and females were combined and when eGFR was examined as a function of titer/day
Breunig 2006 [68]25Mean (SD) [range]:
23 (8) [12, 37] months
  • A single-center observational study of German patients with different stages of FD
  • Mean (SD) GFR (mL/min/1.73 m2)
    Patients with GFR > 90 (n = 16): baseline, 115 (18); follow-up, 102 (14); p = 0.15
    Patients with GFR < 90 (n = 16): baseline, 71 (17) [n = 8]; follow-up, 60 (23); p = 0.039
Burlina 2019 * [70]
The Fabry Registry
49≥2.5 years
  • Observational study using data from the Fabry Registry
  • Included patients started on agalsidase beta (1 mg/kg EOW) when aged 5–30 years
  • eGFR outcomes were stratified by renal involvement (high and low) based on albuminuria/proteinuria levels
  • eGFR decline was higher among 16 high renal involvement males than 66 LRI males (slopes, −2.56 vs. −1.29 mL/min/1.73 m2/year; p value of interaction <0.05; median age at first treatment 25.3 vs. 20.6 years)
  • Slope estimates were unreliable among females
Cabrera 2017 [30]39Mean (SD):
68.2 (36.9) months
  • Long-term prospective and historical analysis of cohort of patients with FD from Argentina
  • At baseline, 5 patients had eGFR < 60 mL/min/1.73 m2, 2 males; 3 females
  • Mean (SD), eGFR (mL/min/1.73 m2):
    Males (n = 17): baseline: 96.9 (28); follow-up: 102 (29)
    Females (n = 13): baseline: 86 (23); follow-up: 84 (23)
Chen 2024 * [83]2248 weeks
  • Phase 4 open-label study of Chinese patients
  • Baseline mean (SD) eGFR: 115.6 (37.3) mL/min/1.73 m2
  • Week 48 mean (SD) eGFR: 118.4 (42.9) mL/min/1.73 m2
  • Change: 0.0 (12.4) mL/min/1.73 m2
Dutra-Clarke 2021 [71]202–20 years
  • Observational study in the US
  • Annual decrease in eGFR of −1.88 mL/min/1.73 m2 (95% CI: −3.36, −0.40; p = 0.013) on ERT in males and females and −3.16 mL/min/1.73 m2 (95% CI: −4.97, −1.35; p = 0.001) for males alone
Germain 2015 [82]52Median [IQR]:
10 [7.3, 10.3] years
  • Analysis of phase 3 clinical trial data and Fabry Registry observational analysis
  • LRI subgroup (n= 32): mean eGFR slope: −1.89 mL/min/1.73 m2/year
    p = 0.001
  • HRI subgroup (n = 20): mean eGFR slope: −6.82 mL/min/1.73 m2/year
    p < 0.001
  • Patients with LRI whose UPCR was ≤0.5 g/g throughout the treatment period: mean slope: −1.48 mL/min/1.73 m2/year p < 0.001
  • Patients with LRI whose UPCR rose above 0.5 g/g during the treatment period: mean slope: −2.6 mL/min/1.73 m2/year p < 0.001
Goicoechea 2020 * [72]69Median [range] follow-up:
60 [24, 120] months
  • An observational study of Spanish patients
  • At the end of follow-up, eGFR remained stable in 42 males and 27 females
  • Renal, neurological, or cardiac events were more frequent in patients with baseline eGFR ≤ 60 mL/min/1.73 m2 (log-rank, 12.423;
    p = 0.001), and this remained significant even after excluding incident renal events (log-rank, 4.086; p = 0.043)
  • Lower baseline eGFR was associated with a 3- to 7-fold increase in the risk of clinical events in different Cox models
Hopkin 2023 [23]
The Fabry Registry
785Median:
6.3 years in males;
5.0 years in females
  • Observational study using data from the Fabry Registry
  • Baseline eGFR: males: median 91.7 mL/min/1.73 m2; females: median 102.3 mL/min/1.73 m2; p < 0.001
  • Male patients (n = 524): subgroup with eGFR assessments for slope analysis and renal involvement classification (n = 140); LRI, 117 (83.6%); HRI, 23 (16.4%)
  • eGFR slopes in males
    LRI subgroup: slope, −1.18 mL/min/1.73 m2/year; 95% CI: −1.66, −0.71; p < 0.001
    HRI subgroup: slope, −2.39 mL/min/1.73 m2/year; 95% CI: −3.53, −1.25; p < 0.001
    p difference: 0.055
    Median follow-up: 6.3 years (LRI) vs. 5.6 years (HRI)
  • Female patients (n = 261)
    Subgroup with LRI and eGFR assessments for slope analysis (n = 59); HRI subgroup analysis not possible
    eGFR slope: −0.92 mL/min/1.73 m2/year; 95% CI: −1.81, −0.04; p = 0.040
    Median follow-up: 5.0 years
  • Patients with baseline eGFR > 135 mL/min/1.73 m2
    Male LRI (n = 7): eGFR slope, −2.95 mL/min/1.73 m2/year; 95% CI: −6.01, 0.12; p = 0.057
    Female LRI (n = 6): eGFR slope, −3.20 mL/min/1.73 m2/year; 95% CI: −9.71, 3.32; p = 0.140
    p difference: 0.893
    Median follow-up: 4.9 years (males) vs. 5.8 years (females)
    Median baseline eGFR: 148.4 mL/min/1.73 m2 (males) vs. 138.0 mL/min/1.73 m2 (females)
    Median age at first treatment: 15.5 years (males) vs. 12.9 years (females)
    Median age at last eGFR assessment: 23.6 years (males) vs. 28.4 years (females)
Hwang 2022 [73]1022-week follow-up
Mean (SD) [range]:
4.6 (3.2) [1, 9] years
  • A phase 2 open-label study in Korea
  • 8 patients had received previous ERT with agalsidase beta, 2 patients were treatment naive
  • Mean (SD) [range] eGFR: baseline: 107.00 (28.42) [68.00, 159.00] mL/min/1.73 m2; follow-up: 116.33 (29.77) [77.00–163.00] mL/min/m2
Lubanda 2009 [74]2196 weeks
  • Evaluation of a low dose, after a standard therapeutic dose, of agalsidase beta
  • Patients received 12 infusions of agalsidase beta at 1.0 mg/kg EOW for 6 months (weeks 0–23), followed by up to 37 infusions at 0.3 mg/kg EOW for 18 months (weeks 24–96)
    Mean, median (SD) [range] eGFR (mL/min/1.73 m2): baseline:
    (n = 21), 93, 92.5 (21.0) [55–134]
    Median eGFR (mL/min/1.73 m2): 24 weeks, 93.1; 96 weeks, 92.8
Ortiz 2021 * [75]254Median post-treatment period: 4.1 years
  • Self-controlled comparison of renal function before and after ERT in male patients with LRI versus HRI
  • LRI (UPCR ≤ 0.5 g/g or UACR ≤ 0.3 g/g) and HRI (ratios > 0.5 g/g or >0.3 g/g, respectively)
    Pre-treatment eGFR slope (mL/min/1.73 m2/year): whole cohort (N = 254): −2.22 (median pre-treatment period 1.1 years); LRI (n = 251): −1.73; HRI (N = 68): −2.93
    eGFR slope, pre- vs. post-treatment (mL/min/1.73 m2/year): whole cohort (n = 254), −2.22 vs. −2.66 (p pre-post difference = 0.24; median pre-treatment period, 1.1 years; median post-treatment period, 4.1 years); LRI (n = 165), −1.73 vs. −1.92 (p pre-post difference = 0.66); HRI (n = 68), −2.93 vs. −4.31 (p pre-post difference = 0.04); statistically significant between LRI and HRI, p interaction <0.01
Ramaswami 2019 [76]315 years
  • Pediatric male patients treated with 2 low-dose regimens (0.5 mg/kg q2w, n = 16; or 1.0 mg/kg q4w, n = 15)
  • Mean, median (SD) [range] iGFR (mL/min/1.73 m2)
    Year 5 overall (n = 23), 115.7, 112.4 (15.4) [90.6, 149.1]; change from baseline to year 5, −3.4, 0.5 (12.7) [−35.9, 19.2], p = 0.4065; annualized rate of iGFR decline, −0.7, 0.1 (2.5) [−7.2, 3.9]; % change from baseline, −2.0, 0.3 (10.5) [−27.4, 18.7]
  • Mean, median (SD) [range] eGFR (mL/min/1.73 m2)
    Year 5 overall (n = 27), 118.6, 121.0 (21.6) [81.1, 168.7]; change from baseline to year 5, −0.7, −4.6 (17.1) [−25.4, 58.3], p = 0.2667; annualized rate of eGFR decline, −0.1, −0.9 (3.4) [−5.1, 1.7]; % change from baseline, 0.8, −3.5 (16.4) [−20.1, 56.7]
Veloso 2023 * [69]9Mean (SD):
70.2 (12.9) months
  • Follow-up study of a family with a classic pathogenic variant (p.G35V) in Brazil
  • eGFR and proteinuria/albuminuria remained stable during the follow-up in both sexes (further details not reported in abstract)
Wanner 2020 [77]
Fabry Registry
86Mean (SD) [range]:
3.9 (0.9)
[2.0, 5.0] years
  • Self-controlled pretreatment and post-treatment comparison in adult female patients in the Fabry Registry
  • Pretreatment vs. post-treatment eGFR slopes (95% CI) mL/min/1.73 m2/year overall and stratified by renal involvement or use of ACEI/ARBs
    Overall (n = 86): −0.83 (−1.52, −0.13) during pre-treatment and −0.95 (−1.59, −0.32) during post-treatment
    HRI (n = 22): eGFR slope pretreatment (95% CI), −1.95 (−3.23, −0.67), p < 0.01, p (pre-difference) = 0.10; eGFR slope post-treatment (95% CI), 2.08 (−3.21, −0.95), p < 0.01, p (post-difference) < 0.01; pretreatment vs. post-treatment eGFR slope difference (95% CI), −0.13 (−1.99, 1.72), p (pre-post difference) = 0.89, p (interaction) = 0.58
    LRI (n = 53): eGFR slope pretreatment (95% CI), −0.68 (−1.56, 0.20), p = 0.13; eGFR slope post-treatment (95% CI), −0.14 (−0.92, 0.63), p (post-difference) = 0.72; pretreatment vs. post-treatment slope difference (95% CI), 0.54 (−0.81, 1.88), p (pre-post difference) = 0.43)
    ‘Ever’ ACEI/ARBs (n = 52): eGFR slope pretreatment (95% CI), −1.30 (−2.17, −0.43), p (pre-difference) = 0.08; eGFR slope post-treatment (95% CI), −1.19 (−1.96, −0.43), p = < 0.01, p (post-difference) 0.38; pretreatment vs. post-treatment eGFR slope difference (95% CI), 0.10 (−1.13, 1.34), p (pre-post difference) 0.87, p (interaction) = 0.55
    ‘Never’ ACEI/ARBs (n = 34): eGFR slope pretreatment (95% CI), 0.01 (−1.15, 1.18), p = 0.98; slope post-treatment (95% CI), −0.57 (−1.69, 0.56), p = 0.32; pretreatment vs. post-treatment slope difference (95% CI), −0.58 (−2.43, 1.27), p (pre-post difference) = 0.54
Warnock 2012 [78]
Fabry Registry
213≥2 years
  • Observational study using data from the Fabry Registry
  • Multivariate logistic regression analyses to identify factors associated with renal disease progression
  • Patients were categorized into quartiles based on eGFR slopes during treatment.
    Men’s eGFR slope (mL/min/1.73 m2/year) categories: quartile 1, −1.2 to 15.3; quartile 2, −2.8 to 1.3; quartile 3, −4.8 to 2.9; and quartile 4, −15.5 to −14.9.
    Women’s eGFR slope (mL/min/1.73 m2/year) categories: quartile 1, 1.1 to 7.5; quartile 2, −0.4 to 1.0; quartile 3, −2.3 to −0.4; and quartile 4, −7.6 to −2.4
  • Mean, median (SD) [IQR] pre-treatment (baseline) eGFR (mL/min/1.73 m2)
    Men: eGFR slope quartile 1 (n = 37), 99, 104 (22.6) [89, 115]; eGFR slope quartile 2 (n = 37), 77, 73 (32.9) [46, 108]; eGFR slope quartile 3 (n = 38), 88, 88 (38.7) [51, 123]; eGFR slope quartile 4 (n = 36), 72, 61 (34.3) [47, 95]
    Women: eGFR slope quartile 1 (n = 14): 88, 87 (16.0) [75, 100]; eGFR slope quartile 2 (n = 14): 90, 93 (23.9) [72, 111]; eGFR slope quartile 3 (n = 16): 95, 98 (27.6) [76, 118]; eGFR slope quartile 4 (n = 14): 78, 85 (35.6) [36, 105]
  • Mean, median (SD) [IQR] eGFR slope (mL/min/1.73 m2/year)
    Men: eGFR slope quartile 1 (n = 38): −0.1, −0.5 (1.20) [−0.9, 0.6]; eGFR slope quartile 2 (n = 38), −2.1, −2.1 (0.52) [−2.6, −1.5]; eGFR slope quartile 3 (n = 38), −3.8, −3.6 (0.61) [−4.3, −3.3]; eGFR slope quartile 4 (n = 37), −6.7, −6.2 (2.26) [−6.9, −5.5]
    Women: eGFR slope quartile 1 (n = 15), 2.7, 2.6 (1.65) [1.3– 3.4]; eGFR slope quartile 2 (n = 16), 0.1, 0.1 (0.41) [−0.2, 0.4]; eGFR slope quartile 3 (n = 16), −1.3,−1.4 (0.66) [−1.7, −0.6]; eGFR slope quartile 4 (n = 15), −4.4, −4.2 (1.58) [−5.5, −3.0]
  • The risk factor most strongly associated with renal disease progression was UPCR ≥ 1 g/g (OR: 112; 95% CI: 4, 3109; p = 0.0054)
  • Longer time from symptom onset to treatment was also associated with renal disease progression (OR: 19; 95% CI: 2, 184; p = 0.0098)
Warnock 2015 [79]
FAACET
24Median [IQR] duration of therapy before first visit:
3.1 [0.3, 4.4] years
Patients were followed up during a 21-month treatment phase
  • The FAACET study evaluated the safety and efficacy of proteinuria control with ACEI and/or ARB therapy in patients with FD receiving ERT with agalsidase beta
  • Assessment of renal function in patients with classic FD stratified by achieved UPCR goal (defined as UPCR for first visit ≤ 0.5 g/g or averaged treatment UPCR ≤ 50% of baseline or averaged treatment UPCR ≤ 0.5 g/g)
  • eGFR slope (mL/min/1.73 m2/year)
    Patients who met UPCR goal (n = 18), eGFR slope (95% CI) −3.6 (−4.8, −1.1)
    Patients who did not meet UPCR goal (n = 6), eGFR slope (95% CI) −7.0 (−5.6, −2.0)
  • Median [IQR] eGFR at initial follow-up visit (mL/min/1.73 m2):
    Patients who met UPCR goal and eGFR slope ≥ −2 mL/min/1.73 m2/year (n = 6): 88 [56, 11]
    Patients who met UPCR goal and eGFR slope < −2 mL/min/1.73 m2/year (n = 12): 62 [48, 78]
    Patients who were above UPCR goal and eGFR slope < −2 mL/min/1.73 m2/year; (n = 6), 61 [45, 81]
  • Median (IQR) eGFR—average of follow-up visits (mL/min/1.73 m2):
    Patients who met UPCR goal and eGFR slope ≥ −2 mL/min/1.73 m2/year (n = 6): 80 [75, 126]
    Patients who met UPCR goal and eGFR slope < −2 mL/min/1.73 m2/year (n = 12): 56 [45, 75]
    Patients who were above UPCR goal and eGFR slope < −2 mL/min/1.73 m2/year (n = 6): 68 [45, 73]
  • Median (IQR) eGFR at last follow-up visit (mL/min/1.73 m2):
    Patients who met UPCR goal and eGFR slope ≥ −2 mL/min/1.73 m2/year (n = 6): 97 [75, 121]
    Patients who met the UPCR goal (and eGFR slope < −2 mL/min/1.73 m2/year (n = 12): 62 [51, 85]
    Patients who were above UPCR goal and eGFR slope < −2 mL/min/1.73 m2/year (n = 6): 47 [35, 90]
Weidemann 2013 [80]40Median [IQR]: 6.0 [5.1, 7.2] years
  • Assessment of long-term effects of agalsidase beta in patients with advanced FD at a single center in Germany
  • Mean (SD) GFR (mL/min): baseline, 85 (28); at follow-up, 73 (39) (p = 0.003)
  • Mean (SD) annual change in GFR −2.3 (4.6) mL/min/year (men: −2.4 (5.0) mL/min/1.73 m2/year and women: −1.9 (3.3) mL/min/year)
Wraith 2008 [81]1648 weeks
  • Open-label study of pediatric patients with FD from France, Poland UK, and USA
  • Mean (SD) eGFR (mL/min/1.73 m2): baseline (n = 16), 126 (29); week 24 (n = 16), 128 (29); week 48 (n = 15), 125 (26)
(B)
Author Year
Study Identifier
Treatment Groups, n
Treatment Duration
Key Results
Comparator studies
Arends 2018 [29]Agalsidase alfa: 248
Agalsidase beta: 139
Median [range]:
All patients
4.9 [0.8, 14.4] years;
Agalsidase alfa
5.2 [0.8. 14.4] years;
Agalsidase beta
3.8 [0.8, 12.1] years
  • Retrospective cohort study involving three European centers combined with data from the Canadian FD Initiative
  • Median eGFR [range], mL/min/1.73 m2 at baseline: agalsidase alfa, 89 [10, 159]; agalsidase beta, 86 [10, 140]
  • Adjusted for sex and phenotype, there was no difference in eGFR slope between agalsidase alfa and beta in patients with a baseline eGFR ≥ 60 mL/min/1.73 m2 (beta slope alfa-beta: −0.12 mL/min/1.73 m2/year; 95% CI: −0.76, 0.51; p = 0.70)
  • In patients with an eGFR < 60 mL/min/1.73 m2, there was no difference in the rate of decline (beta slope alfa-beta: −0.85 mL/min/1.73 m2/year, 95% CI: −2.31, 0.62; p = 0.26), and adding the use of ACEI/ARBs and/or presence of proteinuria at baseline as covariates to the model did not result in a better fit or different results
Beck 2015 [112]
FOS
Agalsidase alfa: 740
Published findings for untreated patients
Median [range]:
5.4 [1.5, 13.7] years
  • Mean annualized eGFR slope (SEM) [95% CI], mL/min/1.73 m2/year
  • FOS evaluable treated renal cohort by baseline eGFR category (mL/min/1.73 m2):
    Males, ≥ 60 (n = 117): −1.68 (0.19) [−2.05, −1.31]; <60 (n = 18): −2.86 (0.53) [−3.90, −1.83]
    Females, ≥ 60 (n = 111): −0.43 (0.21) [−0.83, −0.02]; <60 (n = 22): 0.36 (0.42) [−0.47, 1.19]
  • Previously published external untreated cohort by baseline eGFR category (mL/min/1.73 m2):
    Males, ≥60 (n = 117): −3.0 (0.1); <60 (n = 28): −6.8 (1.5)
    Females, ≥60 (n = 42): −0.9 (0.9); females, <60: (n = 13); −2.1 (1.6)
  • In a sensitivity analysis, eGFR results were evaluated only for FOS ERT patients who had 5-year data (79 males, 71 females; median [range] treatment duration, 8.25 [2.7, 13.8] years)
    The annualized eGFR slopes for these patients, overall and grouped by baseline eGFR, were not significantly different from corresponding values for the full FOS ERT renal cohort
Cybulla 2009 [113]
FOS
Kidney transplant recipients: 27
(20 received agalsidase alfa, 7 untreated)
Median ERT treatment duration: 3.5 years
  • Baseline renal function data available for 27 kidney transplant recipients (n = 27): median [IQR] eGFR (mL/min/1.73 m2) 44.4 [33.6, 64.0]
  • Renal function in kidney transplant recipients receiving ERT or without ERT, medians [IQR] eGFR (mL/min/1.73 m2):
    Kidney recipients receiving ERT (n = 20), 59.2 [36.5, 65.2]
    Kidney recipients without ERT (n = 7), 35.1 [19.9, 51.7]
Cybulla 2013 * [115]
FOS
Kidney transplant
recipients: 93
(78 treated with ERT before
or after transplant surgery,
15 untreated)
Mean (SD):
3.7 (3.1) years
  • Mean (SD) baseline eGFR (mL/min/1.73 m2)
    Kidney recipients receiving ERT (n = 53): 44.8 (25.9)
    Kidney recipients without ERT (n = 10): 61.7 (12.0)
  • Mean (95% CI) eGFR slope (mL/min/1.73 m2/year)
    Male recipients receiving ERT: −0.0 (−2.7, 2.6)
    Male recipients without ERT: −4.5 (−14.1, 5.0)
    Female recipients receiving ERT: −1.0 (−10.4, 8.4)
    Female recipients without ERT: −2.8 (−24.5, 18.8)
Cybulla 2014 * [114]
FOS
Kidney transplant recipients: 51 (47 treated with ERT before or after transplant surgery, 4 untreated)
Median [range]:
2.9 [0.0, 13.4] years
  • Mean [range] baseline eGFR (mL/min/1.73m2)
    Kidney recipients receiving ERT (n = 24): 45.7 [4.1, 110.8]
    Kidney recipients without ERT (n = 3): 54.2 [45.9, 59.7]
Dehout 2003 [116]
FOS
Agalsidase alfa: 234, Untreated: 103
12 months
  • Observational study using preliminary data from FOS
  • Patients with baseline GFR 60–89 mL/min (n = 39): a significant improvement in GFR from baseline, p = 0.027 following 12 months of treatment with agalsidase alfa
  • Patients with baseline GFR 30–59 mL/min (n = NR): GFR was maintained after 12 months of treatment with agalsidase alfa, whereas in those without treatment, GFR declined significantly (p < 0.021)
  • Patients with a baseline GFR > 90 mL/min (n = 51): no difference between treated and untreated patients after 1 year
Golan 2015 [118]Agalsidase alfa 0.2 mg/kg EOW: 20
Agalsidase alfa 0.2 mg/kg weekly: 19
Agalsidase alfa 0.4 mg/kg weekly: 5
1 year
  • Phase 3/4 multi-center open-label study
  • Mean baseline eGFR (mL/min/1.73 m2)
    0.2 mg/kg EOW (n = 20): 82.0
    0.2 mg/kg weekly (n = 19): 78.1
    0.4 mg/kg weekly (n = 5): 72.1
  • Median baseline UACR
    0.2 mg/kg EOW (n = 20): 41.5
    0.2 mg/kg weekly (n = 19): 40.0
    0.4 mg/kg weekly (n = 5): 250.0
  • Week 53 mean eGFR (mL/min/1.73 m2)
    0.2 mg/kg EOW (n = 17): 78.3
    0.2 mg/kg weekly (n = 18): 77.5
    0.4 mg/kg weekly (n = 5): 70.4
  • Change in eGFR (95% CI) at week 53 (mL/min/1.73 m2)
    0.2 mg/kg EOW: −1.21 (−7.46, 5.05)
    0.2 mg/kg weekly: −3.32 (−9.63, 2.99)
    0.4 mg/kg weekly: −1.68 (−14.00, 10.64)
  • 0.2 mg/kg weekly minus EOW mean eGFR (mL/min/1.73 m2)
    LSM difference: −1.56 (95% CI: −9.23, 6.12); unadjusted p = 0.6830
  • Mean (95% CI) change from baseline in eGFR by CKD stage, 1A, 1B, 2, 3, 4/5 (mL/min/1.73 m2)
    0.2 mg/kg EOW: CKD-1A, −24.2 (−82.6, 34.2; n = 3); CKD-1B, 1.6 (−35.4, 38.7; n = 3); CKD-2, 4.5 (−1.2, 10.1; n = 10); CKD-3, −5.0 (−27.3, 17.2; n = 3); CKD-4/5, −1.2 (−7.5, 5.0; n = 1)
    0.2 mg/kg weekly: CKD-1A, −24.4 (−71.4, 22.6; n = 2); CKD-1B, −4.3 (−21.4, 12.8; n = 5); CKD-2, −1.2 (−8.4, 5.9; n = 7); CKD-3, 7.3 (−24.6, 39.2; n = 3); CKD-4/5, −2.9 (n = 2)
    0.4 mg/kg weekly: CKD-1A, NA (n = 0); CKD-1B, −0.4 (n = 1); CKD-2, −2.3 (−37.0, 32.4; n = 3); CKD-3, NA (n = 0); CKD-4/5, −1.0 (n = 1)
Guerard 2018 [119]Lucerastat + ERT:
10 ERT only: 4
12 weeks
Mean (SD) previous ERT at baseline:
Lucerastat:
4.5 (2.6) years;
ERT only:
6.3 (4.2) years
  • Single-center, open-label, randomized study in Germany
  • Mean (SD) baseline eGFR (mL/min/1.73 m2)
    Lucerastat + ERT (n = 10): 88.7 (26.2)
    ERT only (n = 4): 94.9 (26.7)
  • Week 4 mean (SD) eGFR
    Lucerastat + ERT (n = 10): 97.4 (26.0)
    ERT only (n = 4): 91.8 (20.5)
  • Week 8 mean (SD) eGFR
    Lucerastat + ERT (n = 10): 90.3 (21.6)
    ERT only (n = 4): 88.8 (23.8)
  • Week 12 mean (SD) eGFR
    Lucerastat + ERT (n = 10): 92.9 (23.1)
    ERT only (n = 4): 95.0 (24.1)
Hughes 2008 [121]Agalsidase alfa: 7
Placebo: 8
Randomized phase: 6 months OLE: 2 years
  • Placebo-controlled study followed by open-label extension for 2 years
  • Mean baseline GFR showed a statistically significant increase within the normal range (106–131 mL/min; p = 0.007) in the drug group, whereas the placebo groups did not show any statistically significant change throughout the 6-month study
Hughes 2019 * [120]Agalsidase alfa: 21
Untreated: 52
NR
  • Mean (SD) eGFR changes in women with FD from before to after pregnancy (mL/min/1.73 m2)
    ERT: −1.5 (11.7)
    Untreated: −3.1 (13.6)
Jovanovic
2017 * [122]
FACETS
ATTRACT
FACETS:
Migalastat or
placebo: 67;
OLE, migalastat: 54,
24 months
ATTRACT:
Migalastat or ERT: 52, 18 months
Efficacy analyses focused on patients with amenable mutations, FACETS:
(n = 41); ATTRACT:
(n = 34)
  • Over 24 months in FACETS, the annualized rate of change in eGFR CKD-EPI (SEM) with migalastat was −0.3 (0.664) mL/min/1.73 m2
  • The annualized rate of change (SEM) for eGFR in the range of 60 to <90 mL/min/1.73 m2 was −0.8 (1.01), which was comparable to the rate in the range of ≥90 mL/min/1.73 m2, 0.2 (0.90)
  • During the 18-month controlled period of ATTRACT, the annualized means (SD) for eGFR for migalastat and ERT were comparable, −0.40 (4.3) and −1.03 (7.4), respectively
  • The annualized rates of change in eGFR (SEM) in the range of 60 to <90 mL/min/1.73 m2 for migalastat and ERT were −0.2 (1.25) and −7.3 (6.01), respectively; the annualized rates in the range of ≥90 mL/min/1.73 m2 were −2.0 (0.57) and −2.1 (1.60)
  • During the 12-month OLE, eGFR remained stable
Prabakaran 2014 [123]Agalsidase beta/agalsidase alfa: 13
Untreated: 4
≤6 years
  • Median [range] baseline eGFR (mL/min/1.73 m2)
    ERT-treated: 100 [64, 159]
    Untreated: 99 [88, 124]
  • Mean annualized (95% CI) change in eGFR, (mL/min/1.73 m2)
    ERT-treated: −1.45 (95% CI: −0.31, −2.58; p = 0.012)
    Untreated: −0.89 (95% CI: −1.51, 3.30; p = 0.47)
    Decline in the two groups was not significantly different, p = 0.68
Schiffmann 2018 * [124]Migalastat cohort: 52
Natural history cohort: 90 (untreated patients)
3–5 years
  • Mean annualized (SE) eGFR rate of change (mL/min/1.73 m2) by baseline 24-h urine protein (mg/24 h)
    Migalastat males
    -
    <100 (n = 3): 1.2 (1.2)
    -
    100–1000 (n = 16): 0.9 (1.0)
    -
    1000 (n = 2): −4.3 (0.1)
    Migalastat females
    -
    <100 (n = 9): −0.9 (0.5)
    -
    100–1000 (n = 19): 1.3 (1.5)
    -
    1000 (n = 3): −1.7 (1.1)
    Natural history cohort males
    -
    <100 (n = 18): −1.6 (1.5)
    -
    100–1000 (n = 21): −3.3 (1.8)
    -
    1000 (n = 22): −6.9 (1.5)
    Natural history cohort females
    -
    <100 (n = 7): −0.6 (2.6)
    -
    100–1000 (n = 17): −2.2 (2.2)
    -
    >1 000 (n = 5): −4.6 (2.3)
Sirrs 2018 * [27] Agalsidase alfa 0.2 mg/kg: 76
Agalsidase beta 1 mg/kg: 56
Median [range]:
99 [5, 123] months
  • A prospective multi-center study of ERT in Canada
  • Males: rate of decline of eGFR (mL/min/1.73 m2/year) tended to be lower in patients on agalsidase beta than those on agalsidase alfa: −1.98 vs. −4.15, respectively; p = 0.09
  • Females: no difference in the rate of decline in eGFR was seen between the two treatment groups in females
Skuban 2017 * [108]
FACETS
ATTRACT
FACETS:
Migalastat
or placebo: 67
OLE, migalastat: 60
FACETS: 6-month double-blind followed by open-label migalastat from 6 to 12 months (stage 2) plus an additional year
ATTRACT:
Migalastat or ERT: 53.18 months
  • Analysis of 2 randomized phase 3 clinical studies
  • FACETS: mean annualized rate of change (SEM) over 24 months in eGFR CKD-EPI with migalastat was −0.3 (0.7) mL/min/1.73 m2
  • ATTRACT: migalastat and ERT had comparable effects on renal function during the 18-month controlled period
  • Compared with natural history, the rate of GFR decline in FACETS was slower than that of untreated patients when matched for sex and baseline proteinuria
van der Veen 2022 [33]Agalsidase beta
0.5 mg/kg: 3
Agalsidase beta
1 mg/kg: 4
Untreated: 23
10 years
  • Cross-sectional retrospective study comparing outcomes in patients with classic FD after 10 years of ERT to a group of untreated patients with the same phenotype of comparable age
  • Median [range] eGFR (mL/min/1.73 m2)
    Treated: 116 [92, 132]
    Untreated: 116 [46, 165]; p = 0.96
  • Reduced renal function (eGFR < 90 mL/min/1.73 m2)
    Treated: n = 0
    Untreated: n = 4
  • Hyperfiltration (eGFR > 140 mL/min/1.73 m2)
    Treated: n = 0
    Untreated: n = 3
Vedder 2007 [28]Agalsidase alfa: 18
Agalsidase beta: 16
24 months
  • Median (SD) [range] GFR (mL/min)
    12 months: agalsidase beta, 111 (30) [56, 159]; agalsidase alfa, 102 (42) [16, 150]
    24 months: agalsidase beta, 107 (31) [49, 144]; agalsidase alfa, 100 (40) [14, 158]
    No significant differences between groups or from baseline in either group
Wallace 2022 * [125], Wallace 2024 [127]
NCT02795676
Pegunigalsidase alfa: 52
Agalsidase beta: 25
24 months
  • Median [range] eGFR at baseline (mL/min/1.73 m2)
    Pegunigalsidase alfa: 73.5 [30.2, 125.9]
    Agalsidase beta: 74.9 [34.1, 107.6]
    Overall: 74.5 [30.2, 125.9] (p = 0.82)
  • Median eGFR change from baseline at 24 months (mL/min/1.73 m2)
    Pegunigalsidase alfa: −2.39
    Agalsidase alfa: −3.20
  • Median (95% CI) annual eGFR slope at 24 months (mL/min/1.73 m2/year)
    Pegunigalsidase alfa (n = 52): −2.51 (−3.79, −1.24)
    Agalsidase beta: (n = 25): −2.16 (−3.81, −0)
    Difference: −0.36 (−2.44, 1.73)
    Males
    -
    Pegunigalsidase alfa (n = 28): −3.44 (−5.38, −1.73)
    -
    Agalsidase beta (n = 18): −2. 01 (−3.98, −0.04)
    -
    Difference: −1.43 (−3.96, 1.10)
    Females
    -
    Pegunigalsidase alfa (n = 23): −1.15 (−3.11, 0.81)
    -
    Agalsidase beta (n = 7): −2.79 (−6.28, 0.70)
    -
    Difference: 1.64 (−2.56, 5.84)
Eng 2001 [117], Wilcox 2004 [126]Agalsidase beta: 29
Placebo: 29
Initial trial: 20 weeks
Follow-up: 30–36 months
  • Mean baseline (SD) eGFR (mL/min)
    Agalsidase beta: 83.0 (22.0)
    Placebo: 96.6 (35.5)
  • Mean eGFR at start of extension study (mL/min/1.73 m2)
    Agalsidase beta/agalsidase beta: 109.6
    Placebo/agalsidase beta: 138.3
  • Mean eGFR at 30 months of extension study (mL/min/1.73 m2)
    Agalsidase beta/agalsidase beta: 107.1
    Placebo/agalsidase beta: 129.5
  • Patients with (<90 mL/min/1.73 m2) at baseline (n = 10):
    Mean (SD) eGFR (mL/min/1.73 m2)
    Agalsidase beta: 68.6 (19.7) at baseline; and 79.6 (6.3) at 30–36 months after treatment
    After 30–36 months of agalsidase beta treatment, 7 (70%) of the 10 patients had either stabilized their eGFR values (GFR: ± 20%) or improved
* Congress abstract. ACEI, angiotensin-converting enzyme inhibitor. aGAL, α-galactosidase A. ARB, angiotensin receptor blocker. CI, confidence interval. CKD, chronic kidney disease. CKD-EPI, Chronic Kidney Disease Epidemiology Collaboration. eGFR, estimated glomerular filtration rate. EOW, every other week. ERT, enzyme replacement therapy. ESRD, end-stage renal disease. FD, Fabry disease. FOS, Fabry Outcome Survey. GFR, glomerular filtration rate. HRI, high renal involvement. iGFR, iohexol glomerular filtration rate. IgG, immunoglobulin G. IQR, interquartile range. LRI, low renal involvement. LSM, least-squares mean. MDRD, Modification of Diet in Renal Disease. NA, not applicable. NR, not reported. NS, not significant. OLE, open-label extension. q2w, every 2 weeks. q4w, every 4 weeks. SD, standard deviation. SE, standard error. SEM, standard error of the mean. UACR, urine albumin-to-creatinine ratio. UPCR, urine protein-to-creatinine ratio.

3.2. Cardiac Outcomes

Cardiovascular disease is a leading cause of death in patients with FD. Cardiac manifestations of FD include left ventricular hypertrophy (LVH), diastolic dysfunction, valvular irregularities, and conduction disturbances [1]. Publications reporting LVMI data comprised: single-arm studies of agalsidase alfa (n = 19) [39,40,41,42,46,47,48,50,51,52,55,61,63,65,66,128,141,142,143], agalsidase beta (n = 11) [30,69,73,76,80,144,145,146,147,148,149], mixed or non-specified ERT (n = 19) [31,35,36,85,86,88,89,90,91,93,150,151,152,153,154,155,156,157,158], and other treatments (n = 13) [95,98,99,100,101,103,104,105,106,107,108,111,159], comparator studies (n = 15) [29,33,37,112,118,119,121,160,161,162,163,164,165,166], and switch studies (n = 7) [14,128,129,130,134,135,139,140]. Most of the studies (63 studies in 69 publications) assessed LVMI using echocardiography. An overview of the LVMI data from agalsidase alfa and agalsidase beta single-arm studies and the comparator studies is presented in Table 2. Key results from the other types of studies are provided in Supplementary Table S6.
For the 19 publications reporting LVMI data from agalsidase alfa single-arm studies, treatment/follow-up duration varied widely, from 26 weeks to 13 years (Table 2A). Agalsidase alfa stabilized LVMI (i.e., a non-statistically significant change in LVMI or LVM over the follow-up period) in nine studies [42,46,50,52,55,61,66,128,143] and reduced LVMI in three others [39,63,141]. In addition, five studies showed improvement in or stabilization of LVMI for different subgroups [41,47,48,65,142]. A large post-marketing surveillance study of 495 patients in Japan found that treatment with agalsidase alfa for up to 8 years helped to stabilize LVMI in both females and males [52]. Early initiation of treatment with agalsidase alfa in children and adolescents maintained LVMI at pretreatment levels over several years of ERT [50,51,55,76]. In an analysis of FOS registry data (mean treatment duration: ~11 years), male patients with classic FD who started agalsidase alfa treatment before 25 years of age (n = 32) had a nonsignificant mean (standard error [SE]) annual increase in LVMI of 0.28 (0.30) g/m2.7, whereas those who started treatment at or after 25 years of age (n = 58) had a significant mean (SE) annual LVMI increase of 1.03 (0.21) g/m2.7 (p < 0.0001) [41]. Females with FD responded well to agalsidase alfa, with stable or improved LVMI observed over 4–10 years of treatment [47,48,63,142].
All 11 articles that reported the effects of agalsidase beta on LVMI in single-arm studies described a decrease or stabilization of LVMI over treatment periods ranging from 22 weeks to 5 years [30,69,73,76,80,144,145,146,147,148,149] (Table 2A). Three studies that were designed specifically to evaluate the effects of ERT with agalsidase beta on cardiac manifestations demonstrated statistically significant decreases in LVMI over treatment periods of 3–5 years [69,146,147]. Moreover, significant improvements in left ventricular morphology were seen in both males and females, and regardless of FD severity [147]. Analyses by baseline LVH (LVMI > 134 g/m2 in men and >110 g/m2 in women) showed that LVMI reduced significantly in those with baseline LVH (n = 42) and was stable in those without LVH (n = 24) over a median follow-up period of 3 years [147].
The majority of the single-arm studies (10/17) evaluating the effects of mixed or non-specified ERT showed stabilization of LVMI (Supplementary Table S6), with only three studies (two in Taiwanese patients) and one sub-analysis (adults with LVH) reporting a reduction in LVMI during ERT [31,86,151,155]. Most (10/12) of the single-arm studies for other treatments evaluated migalastat. Migalastat led to regression of LVMI in six studies [98,99,100,103,104,105,111] and stabilization in four studies [101,106,108,159] over treatment periods of 48 weeks to 5 years.
Multiple ERT studies assessing LVMI outcomes included a placebo or untreated control arm [33,37,121,156,161,162,165,166], two of which were meta-analyses of previously published data [162,164] (Table 2B). Reductions in LVMI with ERT were demonstrated in both adults [121,161] and adolescents [33], as well as among treatment-naive and treatment-experienced patients [162]. Only one study did not show a difference in LVMI between patients who received ERT (n = 47; median follow-up: 8 years) or no ERT (n = 19; median follow-up: 6 years) [37]. Early initiation of ERT before cardiac disease progression appeared to result in the best LVMI outcomes [156,166]. A randomized, controlled, open-label study found no differences between the efficacy of agalsidase alfa (n = 18) and agalsidase beta (n = 16) on LVM after 12 or 24 months of treatment [28]. In a retrospective cohort study, both agalsidase alfa and agalsidase beta reduced LVMI after 1 year in patients with LVH at baseline (n = 168), with no significant difference between the treatment groups [29]. A greater proportion of patients treated with agalsidase beta than agalsidase alfa had a decrease in LVMI over that period (79% vs. 62%) [29]. In the placebo-controlled FACETS study (n = 67), reductions in LVMI with migalastat were sustained over 30 months in the open-label extension (mean [95% CI] change from baseline: −17.0 [−26.2, −7.9] g/m2; n = 15), including in patients with baseline LVH [160].
The effect of treatment switching on LVMI was assessed in three clinical trials and two observational studies (Supplementary Table S6). In a prospective clinical trial, LVMI was maintained after 24 months in patients who switched from agalsidase beta to agalsidase alfa (n = 71) [128]. No change in LVMI was observed over 12 months in men (n = 15) who switched from agalsidase alfa to pegunigalsidase alfa in the phase 3 BRIDGE trial [139]. In the phase 3 ATTRACT study (n = 36), switching from ERT to migalastat was associated with a mean (95% CI) change in LVMI of −6.6 (−11.0, −2.2) g/m2 after 18 months of treatment [129,130]. However, the inclusion of a broad patient age range (18–72 years) with mild GLA variants associated with residual enzyme activity, may have influenced these results because some patients in this population would not have developed early-onset or severe disease symptoms [167].
Identified studies that assessed other cardiac structure outcomes (e.g., left ventricular posterior wall thickness, interventricular septal thickness, and left ventricular wall thickness) in patients receiving FD treatments are shown in Supplementary Table S7. These included single-arm studies of agalsidase alfa (n = 6) [39,42,46,47,48,61], agalsidase beta (n = 13) [70,73,75,76,77,80,82,144,145,146,147,149,168], mixed or non-specified ERT (n = 9) [85,86,88,89,93,132,152,153,157,163], and other treatments (n = 3, all on migalastat) [98,135,159]. There were seven publications of comparator studies [37,156,158,161,165,169,170] and three publications describing switch studies [129,131,140]. Overall, ERT stabilized cardiac structural changes associated with FD and improved outcomes in patients with LVH. As observed for LVMI, early treatment initiation was more effective at preventing progression than interventions during established disease.
Table 2. Overview of LVMI data from (A) single-arm agalsidase alfa or agalsidase beta studies and (B) comparator studies.
Table 2. Overview of LVMI data from (A) single-arm agalsidase alfa or agalsidase beta studies and (B) comparator studies.
(A)
Author YearNTreatment Duration
Assessment Method
Key Results
Agalsidase alfa single-arm studies
Baehner 2003 [141]15Up to 55 weeks
Echocardiography
  • In heterozygous female patients with FD, mean (SEM) change from baseline in LVMI (g/m2)
    Week 13 (n = 15): −5.5 (5.94); p = 0.372
    Week 27 (n = 11): −23.0 (5.78); p = 0.003
    Week 41 (n = 7): −25.2 (8.12); p = 0.021
Beck 2004 [39]188Mean [max]:
17 [56] months
Echocardiography
  • In a population of patients with FD who were 52% male, echocardiographic data were available for 52/188 (28%) patients treated for 1 year and for 17/92 (18%) patients treated for 2 years
  • Individual data on mean LVM from serial echocardiographic examinations of patients with LVH (LVM > 50 g/m2.7) indicate a decrease in height-adjusted LVM, particularly in those patients with the greatest degree of hypertrophy
  • Overall, there was a significant (p < 0.05) decrease in LVM at 1 and 2 years of treatment with agalsidase alfa
Giugliani 2023 * [40]66Median [IQR]:
20.0 [19.6, 20.6]
NR
  • Observational study using data from FOS
  • Mean (SD) baseline LVMI: 49.79 (14.23) g/m2.7
  • Mean (SE) annual rate of change (n = 31): 0.83 (0.32) g/m2.7
  • Annual increase
    Males: 0.65 g/m2.7
    Females: 1.34 g/m2.7
Goker-Alpan 2015 [128]10024 months
Echocardiography
  • A large cohort study of patients who were started on or switched to agalsidase alfa during a worldwide shortage of agalsidase beta
  • Number of patients with or without baseline LVH
    Naive to ERT (n = 29): with LVH, 10 (34.5%); without LVH, 15 (51.7%); not determined, 4 (13.8%)
    Switch from agalsidase beta to agalsidase alfa (n = 71): with LVH, 27 (38.0%); without LVH, 25 (35.2%); not determined, 19 (26.8%)
  • Mean LVMI did not change significantly in any patient subgroup, including those with or without LVH at baseline
  • Cardiac data presented in figures
Goker-Alpan 2016 [46]14Median [range]:
54.5 [54.0, 59.0] weeks
Echocardiography
  • An open-label safety study of agalsidase alfa in children naive to ERT
  • LVMI values remained within the normal range throughout the study
  • Mean [95% CI] change from baseline at week 55
    Males (n = 5): −0.74 [−6.01, 4.53] g/m2.7
    Females (n = 9): 0.66 [−4.77, 6.08] g/m2.7
Hughes 2009 * [142]
FOS
2504 years
Echocardiography
  • Observational study using data from FOS
  • Cohort of females (n = 78) and males (n = 172) followed for 4 years of ERT
  • After 4 years of agalsidase alfa, there was a significant decrease (p = 0.0312) in mean LVM in females (n = 24), which was not apparent in males (n = 45)
Hughes 2011 [48]
FOS
250≥4 years
Echocardiography
  • Observational study using data from FOS
  • Mean (SD) LVMI (g/m2.7) at 4 years
    Females (n = 24): 43.7 (14.3); p = 0.031 vs. baseline
    Males (n = 45): 52.2 (19.2); p = 0.247 vs. baseline
  • Males with LVH at baseline (n = 22): mean difference after 4 years, −9.11 g/m2.7; p = 0.0115
  • Males with normal LVMI at baseline (n = 23): mean difference after 4 years, −3.82 g/m2.7; p = 0.0741
Kampmann, 2009 [143]4530–42 months
(defined as 36 months)
Echocardiography
  • Retrospective, blinded, pooled analysis of 4 industry-sponsored clinical studies including patients with FD who had received≥ 36 months of ERT
  • LVH was present in 14/45 patients at baseline
  • LVMI (g/m2.7) at 12 months in patients with LVH at baseline (n = 9):
    Mean (SD) change from baseline: −9.2 (7.9)
    Median change from baseline: −8.9, p vs. baseline: 0.008
  • LVMI (g/m2.7) at 36 months in patients with LVH at baseline (n = 10):
    Mean (SD) change from baseline: −5.1 (7.5)
    Median change from baseline: −9.0, p vs. baseline: 0.037
  • LVMI (g/m2.7) at 12 months in patients without LVH at baseline (n = 28):
    Mean (SD) change from baseline: 3.6 (5.7)
    Median change from baseline: 3.2, p vs. baseline: 0.002
  • LVMI (g/m2.7) at 36 months in patients without LVH at baseline (n = 26):
    Mean (SD) change from baseline: 2.1 (7.9)
    Median change from baseline: 1.5, p vs. baseline: 0.27
Kampmann 2015 [47]45Median [range]:
10.8 [9.6, 12.5] years
Echocardiography
  • A single-center observational study in Germany
  • At start of treatment, LVMI values suggested a varying degree of LVH (LVMI ≥ 50 g/m2.7)
    Men: 71% (n = 15/21)
    Women: 67% (n = 16/24)
  • After 1 year of treatment
    Males with LVH: marked improvement in LVMI; LS mean [95% CI] change, −16.46 [−23.81, −9.11] g/m2.7; p < 0.0001
    Females with LVH: LS mean [95% CI] change, −16.69 [−23.62, −9.75] g/m2.7; p < 0.0001; sustained for 3 years and no longer significantly different from baseline after 10 years
  • After 10 years of treatment
    Patients without baseline LVH: no change from baseline
    Males with LVH: LVMI was significantly reduced; LS mean [95% CI] change from baseline, −13.55 [−23.05, −4.06] g/m2.7; p = 0.0061
    Females with LVH: LVMI was not significantly changed
Mehta 2009 [65]
FOS
181Mean (SD):
3.1 (2.1) years
Echocardiography
  • Observational study using data from FOS
  • Patients with baseline LVH
  • Year 1 (n = 23):
    Mean baseline LVMI: 68.5 (20.0) g/m2.7
    Mean LVMI at year end: 58.4 (19.6) g/m2.7
    Mean change from baseline: − 10.1 (12.0) g/m2.7, p = 0.0006
  • Year 2 (n = 24)
    Mean baseline LVMI: 70.1 (19.7) g/m2.7
    Mean LVMI at year end: 56.9 (16.7) g/m2.7
    Mean change from baseline: −13.3 (17.2) g/m2.7, p = 0.001
  • Year 3 (n = 29):
    Mean baseline LVMI: 70.1 (21.2) g/m2.7
    Mean LVMI at year end: 59.8 (18.0) g/m2.7
    Mean change from baseline: −10.3 (20.8) g/m2.7, p = 0.0121
  • Year 5 (n = 32):
    Mean baseline LVMI: 71.4 (22.5) g/m2.7
    Mean LVMI at year end: 64.1 (18.7) g/m2.7
    Mean change from baseline: −7.3 (15.3) g/m2.7, p = 0.0111
  • Patients without baseline LVH
  • Year 1 (n = 17):
    Mean baseline LVMI: 38.5 (6.5) g/m2.7
    Mean LVMI at year end: 35.0 (5.7) g/m2.7
  • Mean change from baseline: −3.5 (6.8) g/m2.7, p = NS Year 2 (n = 18):
    Mean baseline LVMI: 38.9 (6.5) g/m2.7
    Mean LVMI at year end: 35.6 (7.4) g/m2.7
    Mean change from baseline: −3.4 (9.2) g/m2.7, p = NS
  • Year 3 (n = 24):
    Mean baseline LVMI: 38.4 (6.2) g/m2.7
    Mean LVMI at year end: 36.9 (6.9) g/m2.7
    Mean change from baseline: −1.5 (8.5) g/m2.7, p = NS
  • Year 5 (n = 25):
    Mean baseline LVMI: 38.7 (6.3) g/m2.7
    Mean LVMI at year end: 40.5 (10.0) g/m2.7
    Mean change from baseline: +1.8 (10.1) g/m2.7, p = NS
Pintos-Morell 2023 [41]
FOS
285Mean (SD):
Early initiators group:
11.2 (5.7);
Late initiators group:
11.8 (5.8)
NR
  • Observational study using data from FOS
  • Annual LVMI increase
    Early-initiators group (n = 32):
    -
    Mean (SE) increase: 0.28 (0.30) g/m2.7/year,
    p = 0.3457
    Late-initiators group (n = 58):
    -
    Mean (SE) increase: 1.03 (0.21) g/m2.7/year,
    p < 0.0001
  • The rate of LVMI change was significantly different between the groups (p = 0.0419)
Ramaswami 2011 [50]
FOS
8Mean (SD):
4.2 (1.9) years
Echocardiography
  • Observational study in pediatric patients using data from FOS
  • Patients received their first dose of agalsidase alfa before reaching the age of 7 years
  • Mean (SD) [range] LVMI (g/m2.7)
    Baseline (n = 6): 40.6 (6.51) [33.3, 52.3]
    At follow-up (n = 6): 31.8 (7.89) [22.4, 45.1]
Ramaswami 2012 [66]
FOS
98≥6 months
Echocardiography
  • Observational study in pediatric patients (<18 years old) using data from FOS
  • 12 months (n = 26): no change in LVMI observed in 26 children
  • 24 months (n = 17): mean (SD) LVMI declined from 35.7 (5.7) at baseline g/m2.7 to 31.7 (5.6) g/m2.7
Ramaswami 2019 [42]
FOS
69Median [range] time from start of ERT to FOS data extraction (years):
Evaluable treated cardiac cohort
(n = 69):
13.6 [10.1–17.1];
Females (n = 34):
12.8 [10.1–16.5];
Males (n = 35):
14.3 [10.2–17.1]
Echocardiography
  • Observational study using data from FOS
  • Number of patients (%) with LVH (defined as LVMI > 48 g/m2.7 in females and LVMI > 50 g/m2.7 in males)
    Evaluable treated cardiac cohort: 32/69 (46.4%)
    Females: 18/34 (52.9%)
    Males: 14/35 (40.0%)
  • Mean (SD) [95% CI] LVMI (g/m2.7)
    At baseline
    -
    Evaluable treated cardiac cohort (n = 69): 50.65 (16.90)
    -
    Females (n = 34): 52.21 (16.99) [46.3–58.1]
    -
    Males (n = 35): 49.13 (16.92) [43.3–54.9]
    At year 10
    -
    Females (n = 34): 58.8 [48.7–68.9]
    -
    Males (n = 35): 52.0 [43.2–60.8]
  • Mean [95% CI] LVMI slope (g/m2.7/year)
    Patients without LVH at baseline
    -
    Females (n = 16): 0.52 [−0.13, 1.17]
    -
    Males (n = 21): 0.57 [0.02, 1.13]
    Patients with LVH at baseline
    -
    Females (n = 18): 1.51 [0.91, 2.12]
    -
    Males (n = 14): 0.87 [0.19, 1.55]
  • Mean [95% CI] LVMI (g/m2.7) among cardiac cohort patients without renal involvement (eGFR > 90 mL/min/1.73 m2 and urinary protein <0.5 g/day) at baseline
    At baseline
    -
    Females (n = 6): 43.3 [27.1, 59.5]
    -
    Males (n = 10): 39.3 [34.1, 44.5]
    At year 10
    -
    Females (n = 6): 45.9 [22.9, 69.0]
    -
    Males (n = 10): 43.2 [36.3, 50.1]
Ries 2006 [51]2426 weeks
Echocardiography
  • Open-label multi-center study in pediatric patients
  • Pediatric patients naive to ERT
  • Mean (SEM) LVMI (g/m2.7) at baseline
    Boys (n = 19): 32.4 (1.3)
    Girls (n = 5): 36.0 (4.0)
  • Mean (SEM) LVMI (g/m2.7) after 25 weeks of treatment
    Boys: 31.4 (1.4)
    Girls: 32.8 (2.3), p vs. baseline NS
  • Children (n = 3) with high–normal LVMI at baseline (>40 g/m2.7) had a mean decrease of 15% in LVM/h after 25 weeks of treatment
Sasa 2019 [52]493Mean [range]:
3.5 [0.0, 7.9] years
Echocardiography
  • A post-marketing surveillance study involving patients with FD in Japan
  • Mean (SD) LVMI at each year, mean (SD) change in LVMI (g/m2.7) from baseline p versus baseline
    Males (classic phenotype)
    -
    Year 0.5 (n = 43): 69.1 (45.7), 1.6 (13.6), p = 0.459
    -
    Year 1 (n = 40): 64.9 (40.0), 0.6 (14.3), p = 0.777
    -
    Year 2 (n = 35): 66.8 (42.2), 3.4 (20.0), p = 0.320
    -
    Year 3 (n = 31): 69.8 (42.1), 3.3 (18.5), p = 0.328
    -
    Year 4 (n = 23): 61.8 (38.2), 7.4 (15.7), p = 0.035
    -
    Year 5 (n = 8): 68.9 (50.9), 15.0 (23.4), p = 0.112
    -
    Year 6 (n = 4): 81.3 (73.2), 23.5 (34.7), p = 0.268
    -
    Year 7 (n = 2): 122.8 (121.8), 51.7 (58.7), p = 0.430
    -
    Year 8 (n = 1): 192.1 (NA), 76.4 (NA)
    Males (non-typical variant)
    -
    Year 0.5 (n = 23): 79.4 (39.6), −1.4 (13.9), p = 0.638
    -
    Year 1 (n = 19): 79.9 (35.7), −5.3 (9.9), p = 0.031
    -
    Year 2 (n = 15): 70.3 (26.7), −8.5 (16.1), p = 0.060
    -
    Year 3 (n = 14): 68.8 (37.3), −4.2 (13.8), p = 0.271
    -
    Year 4 (n = 14): 67.2 (33.2), −3.4 (10.3), p = 0.240
    -
    Year 5 (n = 5): 75.4 (23.3), −0.2 (14.4), p = 0.973
    -
    Year 6 (n = 2): 91.0 (63.5), 15.5 (16.5), p = 0.412
    Females
    -
    Year 0.5 (n = 50): 65.9 (22.8), 1.8 (14.5), p = 0.384
    -
    Year 1 (n = 45): 64.2 (25.1), 2.8 (12.7), p = 0.147
    -
    Year 2 (n = 39): 66.9 (25.5), 2.7 (14.0), p = 0.240
    -
    Year 3 (n = 27): 74.3 (27.1), 4.3 (16.4), p = 0.187
    -
    Year 4 (n = 19): 71.3 (30.0), 2.6 (22.7), p = 0.626
    -
    Year 5 (n = 7): 79.8 (34.0), 4.3 (30.8), p = 0.727
Schiffmann 2014 [55]17Mean (SD):
6.5 (0.6) years
Echocardiography
  • Open-label multi-center extension study for pediatric patients who had completed a 26-week study
  • This study represented the longest assessment of ERT in children with FD in a clinical trial setting
  • Pediatric patients with FD (7–17 years of age at study enrollment) received 6 months of 0.2 mg/kg agalsidase alfa
  • This study was divided into two phases, before (phase 1) and after (phase 2) a change in the agalsidase alfa manufacturing process
  • For all 17 patients, baseline mean (SD) [range] LVMI (g/m2.7) measurements were within the normal range (upper limit of normal range: males = 51 g/m2.7; females = 48 g/m2.7): 30.66 (5.96) [22.7, 42.3]
  • Through phase 1, LVMI decreased from baseline (mean change: −3.25 at week 185)
  • LVMI continued to decrease through phase 2 (except week 104)
  • Despite LVMI fluctuations observed in individual patients, levels remained below LVH criteria throughout the study
Tsuboi 2017 [61]
Japan Fabry Research—002
36Median [range]:
62.5 [8, 84] months
Echocardiography
  • Prospective observational study in Japan
  • Cohort included ERT treatment-naive patients and those who were receiving agalsidase alfa 0.2 mg/kg every 2 weeks
  • At 60 months, LVMI had decreased from 50.3 at baseline to 46.6 g/m2.7
Whybra 2009 [63]364 years
Echocardiography
  • Prospective single-center open-label study of adult women in
    Germany
  • A cohort of female patients with FD who were experiencing manifestations of FD and had not been previously treated with ERT
    LVH (defined as LVM ≥ 48 g/m2.7) at baseline (n/N [%]): 25/36 (69.4%)
    For patients with LVH, mean (SD) [range] LVM (g/m2.7) at baseline: 89.4 (42.1) [52.8, 201.1]
  • Number of patients with LVH at baseline (n = 25) with decreases in LVM
    LVM decrease of >20%: 13/25 (52%)
    LVM decreases of 10–20%: 7/25 (28%)
    LVM decreases of <10%: 5/25 (20%)
  • Mean (SD) LVMI (g/m2.7) over time by number of months of treatment and LVM classification (* p < 0.05 vs. baseline; ** p < 0.01 vs. baseline; *** p < 0.001 vs. baseline)
    LVM classification, > 85 (n = 9)
    -
    Month 12: 98.1 (31.1) ***
    -
    Month 24: 98.1 (31.7) ***
    -
    Month 36: 96.3 (34.0) ***
    -
    Month 48: 91.9 (29.7) ***
    LVM classification, > 60–85 (n = 9)
    -
    Month 12: 57.4 (13.3) ***
    -
    Month 24: 56.5 (14.6) ***
    -
    Month 36: 58.2 (13.2) ***
    -
    Month 48: 62.0 (17.7) ***
    LVM classification, > 48–60 (n = 8)
    -
    Month 12: 46.3 (4.7) ***
    -
    Month 24: 44.0 (5.2) ***
    -
    Month 36: 47.0 (2.8) ***
    -
    Month 48: 48.7 (4.1) ***
    LVM classification, <48 (n = 11)
    -
    Month 12: 36.2 (5.8) **
    -
    Month 24: 35.3 (7.8) **
    -
    Month 36: 37.0 (10.1) *
    -
    Month 48: 36.2 (7.1) **
Agalsidase beta single-arm studies
Cabrera 2017 [30]39Mean (SD):
68.2 (36.9) months
Echocardiography
  • Long-term prospective and historical analysis of a cohort from Argentina
  • Mean (SD) LVMI in males slightly increased from 113.2 (45) to 127.4 (64) g/m2
  • 11 male patients had LVH at baseline
  • In 4/11 males, baseline LVMI was reduced from 163.1 (64.7) to 123.4 (49.8) g/m2, 2 patients had a stabilized LVMI and 4 patients had an increased LVMI from 157.9 (32.3) to 261.6 (48.6) g/m2
  • 9 female patients had LVH at baseline, only 1 patient increased their LVMI from 158 to 204 g/m2; all remaining patients reduced or stabilized their LVMI
  • All patients without LVH at baseline remained with a normal LVMI at the end of the study
Elliot 2006 [144]5Mean (SD):
10.1 (2.3) months
Echocardiography
  • Males receiving 1 or 2 mg/kg agalsidase beta EOW
  • LVMI did not change from baseline to follow-up: mean (SD) [range] change, 0.4 (37.8) [−66.3, 26.4] g/m2; p = NS
Hwang 2022 [73]1022-week follow-up
Mean (SD) [range]: 4.6 (3.2) [1, 9] years
Echocardiography
  • A phase 2 open-label study in Korea
  • 8 patients had received previous ERT with agalsidase beta,
    2 patients were treatment naive
  • Mean (SD) LVMI at week 22: 111.66 (61.74) g/m2; change from baseline, 4.00 (13.44); p = 0.625
Kalliokoski 2006 [145]1012 months
Echocardiography
  • Observational study in Finland
  • Mean (SD) LVM (g)
    Before ERT: 275 (28)
    After 12 months of treatment: 292 (35); p = 0.27
  • Mean (SD) LVMI (g/m2)
    Before ERT: 151 (12)
    After 12 months of treatment: 156 (14); p = 0.37
Messalli 2012 [146]1648 months
MRI
  • Observational study in Italy
  • Study specifically on cardiac structures (MRI study)
  • Baseline: mean (SD) LVM baseline (g), 187 (59)
  • LVM: mean (SD), 149 (44) g; p < 0.001 vs. baseline
Motwani, 2012 [147]66Median [range]:
3 [2, 5] years
Echocardiography
  • Observational study in the United Kingdom
  • The overall mean (SD) LVMI was significantly reduced by ERT at follow-up (n = 66): 116 (28) vs. 113 (26) g/m2; p < 0.001
  • On sub-group analysis, this improvement was seen in both sexes and in both mild (FOS-MSSI ≤18) and moderate/severe (FOS-MSSI > 18) disease groups (all p < 0.05)
  • In patients with LVH at baseline (n = 42), mean (SD) LVMI was significantly reduced by ERT 135 (13) vs. 133 (3) g/m2; p < 0.05
  • There was no significant change in mean (SD) LVMI in those patients without baseline LVH (n = 24): 82 (6) vs. 82 (5) g/m2; p > 0.05
Pisani 2005 [148]924-months
Echocardiography
  • Non-randomized, open-label, prospective study in male patients undergoing maintenance dialysis in the context of renal replacement therapy
  • After 24 months of ERT (n = 9), LVMI did not change compared with basal values
  • Pre- vs. post-changes in LVMI
    In the 24 months before ERT, LVMI increased in all patients, and during the 24 months of therapy, progression of LVMI decreased in all except 2 patients
  • Average percentage of increase per year in LVMI was 6% during the 24 months preceding ERT and 3% during the 2 years of treatment
  • This also was shown by the comparison (p = 0.06) of mean (SD) LVMI curve slopes for the pretreatment period 0.98 (0.017) g/height2.7 with those obtained during the treatment period 0.46 (0.960) g/height2.7
Ramaswami 2019 [76]315 years
Echocardiography
  • Open-label parallel-group phase 3b study in male pediatric patients treated with 2 low-dose regimens (0.5 mg/kg q2w, n = 16 or 1.0 mg/kg q4w, n = 15)
  • Mean, median (SD) [range] LVMI, g/m2.7
    Baseline (n = 23): 33.2, 32.0 (5.1) [20.5, 44.1]
    Year 5 (n = 24): 31.4, 31.9 (7.2) [22.1, 53.4]
Veloso 2023 * [69]9Mean (SD):
70.2 (12.9) months
MRI and Echocardiography
  • Follow-up study of a family with FD with a classic pathogenic variant (p.G35V) in Brazil
  • Female patients presented a mean reduction of LVMI of
    9.73 g/m2 (95% CI: −26.9, 6.8); p = NR
Weidemann 2013 [80]40≥5 years
Median [IQR]:
6.0 [5.1, 7.2]
MRI and Echocardiography
  • Assessment of long-term effects of agalsidase beta in Germany
  • Patients treated for at least 5 years with agalsidase beta
  • Mean (SD) LVM (g)
    Baseline: 270 (87)
    At follow-up: 224 (71); p < 0.0001 vs. baseline
Wuest 2011 [149]1413 ± 1 months
Echocardiography
  • Assessment of the effect of ERT on right ventricular cardiac morphology and function
    Baseline: mean (SD) normalized LVM, 102 (26) g/m2
    At follow-up: mean (SD) normalized LVM, 94 (27) g/m2;
    p < 0.05 between baseline and follow-up
(B)
Author YearComparator Arms, n/NTreatment Duration
Assessment Method
Key Results
Comparator studies
Arends 2018 [29]Agalsidase alfa: 248
Agalsidase beta: 139
Median [range]:
All patients:
4.9 [0.8, 14.4] years; Agalsidase alfa:
5.2 [0.8, 14.4] years; Agalsidase beta:
3.8 [0.8, 12.1] years Echocardiography
  • Retrospective cohort study involving three European centers and the Canadian FD Initiative
    In patients without LVH at baseline (n = 110), there was no change in LVMI after 1 year of ERT
    In patients with LVH (n = 168), there was a decrease after 1 year of treatment
  • The magnitude of the decrease depended on the LVMI at baseline (p < 0.001) and was independent of sex and phenotype
    Patients with an LVMI above the reference value but <75 g/m2.7 treated with agalsidase beta showed a larger but non-significant decrease of LVMI over the first year compared with alfa (beta alfa-beta: −3.31 g/m2.7; 95% CI: −6.84, 0.23; p = 0.07), but no difference for the entire group was found (beta alfa-beta: −2.26 g/m2.7; 95% CI: −5.39, 0.87; p = 0.15)
  • The decrease over the first year was followed by stabilization of LVMI in the following years (beta time on ERT: 0.22; p = 0.33)
  • The analysis of the number of patients who showed a decrease in LVMI after 1 year of treatment revealed that treatment with agalsidase beta resulted in a higher proportion of patients with a decrease in LVMI compared with agalsidase alfa (79% vs. 62%) (OR: 2.27; 95% CI: 1.11, 4.86; p = 0.03), adjusted for the LVMI at baseline
Beck 2015 [112]Agalsidase alfa: 164
Previously published untreated cohort: 78
Median [range]: 5.4 [1.5, 13.7] years
Echocardiography
  • Retrospective comparisons with previously published data (agalsidase alfa-treated patients from the FOS registry with well-described cohorts of untreated individuals from previously published studies)
  • Mean annualized LVMI slope (SEM) [95% CI], g/m2.7/year by baseline LVH status
    Evaluable treated cardiac cohort from FOS
    -
    Males
    Total (n = 71): 0.33 (0.10) [0.13, 0.53]
    LVH (n = 29): 0.19 (0.16) [−0.13, 0.50]
    No LVH (n = 42): 0.47 (0.13) [0.22, 0.72]
    -
    Females
    Total (n = 93): 0.48 (0.09) [0.30, 0.66]
    LVH (n = 45): 0.77 (0.14) [0.49, 1.05]
    No LVH (n = 48): 0.19 (0.11) [−0.03, 0.41]
    Previously published external untreated comparator cohort
    -
    Males
    Total (n = 39): 4.07 (1.03)
    LVH (n = 18): 6.59 (8.5)
    No LVH: not available
    -
    Females
    Total (n = 39): 2.31 (0.81)
    LVH (n = 15): 3.77 (7.7)
    No LVH: not available
  • A sensitivity analysis evaluating available 5-year follow-up LVMI FOS data was conducted in 38 males and 40 females (median [range] treatment duration, 8.45 [0.9, 11.7] years)
    The rates of change in LVMI overall and in patients with baseline LVH were not significantly different from the corresponding values for the full FOS ERT cardiac cohort
Figliozzi 2024 [164]445Median [IQR]:
45 [24–58] months
MRI
  • Systematic review and meta-analysis of patients with baseline and follow-up cardiac MRI after ERT
  • Baseline LVM range: 95–270 g
  • LVMI range: 53–176 g/m2
  • Meta-analysis (random-effects model) of LVMI following ERT showed no changes; mean difference: −1 g/m2 (95% CI: −6, 3; 6 studies; 290 patients; I2 = 81%); p < 0.01
  • Meta-analysis of LVM following ERT: minimal decrease (mean difference: −18 g; 95% CI: −33, −3; 7 studies; 107 patients,
    I2 = 96%); p < 0.01
Germain 2013 [166]Untreated group: 48
Agalsidase beta: 115
(11 included in both panels, because they had at least 2 years of LVM data during each observation period)
Mean [range]:
Untreated group, 4.1 [2.1, 12.7] years;
Agalsidase beta, 4.8 [1.8, 9.5] years
Echocardiography
  • Fabry Registry study to document the progression of LVH in male patients
  • Patients with LVH (LVM ≥ 225 g) at baseline, n/N (%)
    Untreated group: 29/48 (60%)
    Agalsidase beta: 66/115 (57%)
  • At follow-up, LVM slope (g/year) (SEM) by age category
    Age 18–29 years
    -
    During untreated period (n = 15): 9.5 (2.36)
    -
    During treatment (n = 31): −3.6 (1.62)
    -
    Difference in LVM slope: −13.0 (2.72); p < 0.0001
    Age 30–39 years
    -
    During untreated period (n = 17): 8.4 (3.55)
    -
    During treatment (n = 44): 2.8 (2.20)
    -
    Difference in LVM slope: −5.6 (4.12); p = 0.1760
    Age 40–49 years
    -
    During untreated period (n = 7): 13.4 (6.63)
    -
    During treatment (n = 23): 3.4 (2.87)
    -
    Difference in LVM slope: −10.0 (7.21); p = 0.1691
    Age ≥ 50 years
    -
    During untreated period (n = 9): 0.4 (9.41)
    -
    During treatment (n = 17): 7.7 (4.48)
    -
    Difference in LVM slope: 7.3 (10.33); p = 0.4843
Golan 2015 [118]Agalsidase alfa 0.2 mg/kg EOW: 20
Agalsidase alfa 0.2 mg/kg weekly: 19
Agalsidase alfa 0.4 mg/kg weekly: 5
1 year
Echocardiography
  • Phase 3/4 multi-center open-label study in treatment-naive adults
  • Evaluated the efficacy and safety of two agalsidase alfa dosing regimens (0.2 mg/kg EOW and 0.2 mg/kg weekly) in a specific population of adults with FD and LVH (defined as > 50 g/m2.7 for males and > 47 g/m2.7 for females)
  • Change from baseline in mean LVMI, g/m2.7
    0.2 mg/kg EOW (n = 15): 3.2
    0.2 mg/kg weekly (n = 18): 0.5
    0.4 mg/kg weekly (n = 5): −10.3
  • % change (95% CI)
    0.2 mg/kg EOW (n = 15): 3.9 (−3.71, 10.14)
    0.2 mg/kg weekly (n = 18): 3.4 (−7.35, 8.32)
    0.4 mg/kg weekly (n = 5): −9.3 (−24.9, 4.3)
  • 0.2 mg/kg weekly minus EOW: LSM, −2.20 (95% CI: −12.26, 7.85); unadjusted p = 0.6585
Guerard 2018 [119]Lucerastat + ERT: 10
ERT only: 4
Lucerastat 1000 mg b.i.d. for 12 weeks
Echocardiography
  • Single-center, open-label randomized study of lucerastat in adult patients in Germany
  • Investigated the safety and pharmacodynamics of lucerastat as oral substrate reduction therapy in patients with FD receiving ERT
  • All patients were on ERT for at least 24 months with no change in dose in the last 6 months
  • Mean (SD) baseline LVMI (g/m2)
    Lucerastat + ERT (n = 10): 111.7 (38.4)
    ERT only (n = 4): 88.6 (17.9)
  • Mean (SD) LVMI (g/m2) at week 4
    Lucerastat + ERT (n = 10): 104.1 (21.5)
    ERT only (n = 4): 95.3 (35.7)
  • Mean (SD) LVMI (g/m2) at week 8
    Lucerastat + ERT (n = 10): 105.9 (24.7)
    ERT only (n = 4): 90.2 (25.1)
  • Mean (SD) LVMI (g/m2) at week 12
    Lucerastat + ERT (n = 10): 104.4 (24.3)
    ERT only (n = 4): 94.3 (24.3)
  • No effect of treatment with lucerastat on LVMI was observed when comparing values obtained at each visit up to 12 weeks with those at baseline
Hughes 2008 [121]Agalsidase alfa: 7
Placebo: 8
Up to 4 years
MRI and Echocardiography
  • Placebo-controlled study followed by open-label extension for 2 years
  • Mean (SEM) LVMI (g/m2) similar at baseline between groups
    Agalsidase alfa: 176 (10)
    Placebo: 186 (18); p = 0.7
  • Randomized phase
    MRI
    -
    Agalsidase alfa: mean decrease in LVM of 11.5 g
    -
    Placebo: mean increase of 21.8 g (p = 0.041 between groups)
    -
    Equated to mean increase in LVMI for the placebo group of 12 g/m2 compared with a decrease in the treated group of 6.4 g/m2 (p = 0.02 vs. placebo)
    Echocardiography
    -
    Placebo: 6.3% increase in LVM 21.5 (20.4) g
    -
    Agalsidase alfa: 6.2% decrease −20.4 (27.2) g; p > 0.05
    Open-label extension
    -
    MRI measurements in the 10 patients who continued to receive agalsidase alfa in the open-label extension phase of the trial demonstrated individual heterogeneity in response
    -
    After 2 years of active treatment mean LVM was not significantly changed (p = 0.18)
  • Subsequent to the trial, 11 patients continued to receive uninterrupted therapy with agalsidase alfa and have been monitored by annual echocardiography
  • After 4 years, 7 patients demonstrated a reduction in LVM while 4 showed increases compared with their pre-treatment baseline
Jovanovic 2017 * [160]
FACETS
ATTRACT
FACETS Migalastat or placebo: 67
ATTRACT Migalastat or ERT: 60
FACETS: up to 24 months (plus OLE)
ATTRACT: up to 30 months (including OLE)
Echocardiography
  • To assess changes in cardiac parameters with long-term migalastat treatment (150 mg once daily; data from 2 phase 3 RCTs) versus placebo (FACETS) or ERT (ATTRACT)
  • FACETS (migalastat vs. placebo)
    After 18 or 24 months of migalastat treatment in FACETS (18 months for patients initially randomized to placebo, 24 months for patients randomized to migalastat), there was a statistically significant mean change from baseline in LVMI −7.7 g/m2; 95% CI: −15.4, −0.01; n = 27
    Among patients who entered the OLE, further reductions were seen month 30/36: −17.0 g/m2; 95% CI: −26.2, −7.9;
    n = 15, including statistically significant changes in patients with LVH at baseline −20.8 g/m2; 95% CI: −37.4, −4.1; n = 11; 82% (9/11) had reductions and 45% (5/11) had normalizations of LVMI
  • ATTRACT vs. ERT
    LVMI was reduced in patients treated with migalastat in the ATTRACT trial
    Mean changes from baseline at 18 months
    -
    Migalastat (n = 31): −6.6 g/m2 (95% CI: −11.0, −2.1)
    -
    ERT (n = 13): −2.0 g/m2 (95% CI: −11.0, 7.0);
    Mean changes from baseline at 30 months
    -
    Migalastat (n = 30): −3.8 g/m2 (95% CI: −8.9, 1.3)
  • Among those with baseline LVH (n = 13), LVMI was reduced by −9.0 g/m2; 85% (11/13) had reductions and 31% (4/13) had normalizations of LVMI and regression of LVH
Kramer 2014 [161]ERT: 57
No ERT: 16
Mean (SD):
4.8 (2.4) years
MRI
  • Observational study of adults in a single center in Germany
  • Investigated the impact of myocardial fibrosis in FD; ERT vs. no ERT (type of ERT not defined)
  • Mean (SD) LVM (g/m2)
    Baseline
    -
    ERT (n = 57): 92 (33)
    -
    p < 0.05 vs. no ERT at baseline
    -
    No ERT (n = 16): 59 (11)
    At follow-up
    -
    ERT (n = 56): 90 (30)
    -
    p < 0.05 vs. no ERT at follow-up
    -
    No ERT (n = 16): 61 (14)
Lee 2022 [162]ERT: 267
No ERT: 285
4.1 years
NR
  • Meta-analysis to characterize patients with FD with LVH, with or without ERT, and to assess the effectiveness of ERT in LVH improvement
  • Patients (either ERT-experienced [agalsidase alfa or agalsidase beta]) or ERT-naive) included from 5 clinical cohort studies and 2 RCTs
  • Additional subgroup analysis for 4 years after ERT (n = 214) and before ERT (n = 228; patients from 3 cohort studies and 1 RCT)
  • LVMI at follow-up
    Difference in means of LVMI between the ERT treatment group and the ERT treatment-naive group: −0.149 [95% CI: −0.431, 0.132]; p = 0.034
    Difference in means of LVMI between the before ERT group and the 4 years after ERT group: −0.448 [95% CI: −0.787, −0.108]; p = 0.037
Madsen 2017 [37]ERT: 47
No ERT: 19
Median [range]: Patients in ERT 8 [0, 12] years;
Patients without ERT 6 [0, 13] years
Echocardiography
  • Progression of cardiac involvement was assessed in a Danish cohort
  • Baseline LVMI (g/m2), median [range]
    ERT (n = 45): 102 [54, 231]
    Non-ERT (n = 17): 89 [50, 271]
  • LVMI (g/m2) at follow-up, median [range]
    ERT (n = 39): 97 [47, 306]
    No ERT (n = 14): 104 [55, 328]
    ERT with cardiac disease (n = 30): 100 [66, 306]
    ERT without cardiac disease (n = 9): 73 [54, 113]
  • Progression in LVMI (g/m2) per year, estimate (SEM)
    ERT: −0.8 (0.6)
    No ERT: 0.2 (0.7)
    ERT with cardiac disease: −1.0 (0.6)
    ERT without cardiac disease: −0.5 (1.2)
Mignani 2008 [163]Dialysis: 17
Transplant: 17
Mean (SD):
Dialysis patients: 45.1 (19.8) months;
Transplant patients:
48.4 (13.2) months
Echocardiography
  • Cross-sectional survey study of patients in Italy with FD and ESRD receiving RRT (dialysis or renal transplant) and ERT
  • ERT comprised agalsidase beta (n = 30) 1 mg/kg or agalsidase alfa
    (n = 3) 0.2 mg/kg
  • Mean baseline LVMI (g/m2)
    Dialysis patients (n = 7): 210.9
    Transplant patients (n = 11): 234.6
  • LVMI percentage change from baseline at year 3
    Dialysis patients (n = 7): 19%
    Transplant patients (n = 11): −6%
Nordin 2019 [156]Pre-ERT group, 20

Established ERT, 18

No ERT, 18
Mean (SD):
1.1 (0.2) years
Median [range]:
Pre-ERT group:
1 year;
Established ERT: 4.2 [1.4, 12.2] years;
No ERT: 1 year
MRI
  • Pre-ERT (initiated ERT, n = 20)
    Overall, there was no change in mean (SD) LVMI: 93 (42) g/m2 at baseline to 92 (40) g/m2 (p = 0.186) over 1 year after ERT initiation
    Among LVH-positive patients, change in mean (SD) LVMI: −2 (3) g/m2; p = 0.048
    Among LVH-negative patients, no change in mean (SD) LVMI: 0 (4) g/m2; p = 0.766
  • Established ERT (advanced stable disease, n = 18)
    Over 1 year, there was no significant difference in LVMI:
    124 (45) g/m2 at baseline to 125 (45) g/m2 (p = 0.070)
    LVH-positive vs. LVH-negative: NA (sample size too small)
  • No ERT (early disease, mostly females, n = 18)
    Over 1 year, there was a small increase in LVMI: 65 (15) g/m2 at baseline to 67 (16) g/m2 (p = 0.005)
    LVH-positive vs. LVH-negative: NA (sample size too small)
  • Interactions between all three groups revealed statistically significant difference in LVMI (p = 0.009; difference for pre-ERT vs. both established ERT and no ERT)
Pogoda 2023 [165]Untreated controls: 30
Migalastat-treated: 20
ERT-treated: 48
Up to 81 months
Echocardiography
  • A prospective observational single-center study in Germany
  • The control group was untreated females with FD who were younger than the treated group
  • LVMI at baseline, median [range] g/m2
    Untreated females: 82.9 [46.6–155.2]
    Migalastat-treated females: 88.2 [48.6–151.3]
    ERT-treated females: 114.4 [64.9–297.8]
    Migalastat-treated males: 138 [76–220]
    ERT-treated males: 130.2 [80.1–396.0]
  • Change per year in LVMI from baseline, median [range] g/m2
    Untreated females: 1.44 [−44.6, 45.6]
    Migalastat-treated females: 2.57 [−36.8, 10.0]
    ERT-treated females: 0.86 [−10.9, 13.9]
    Migalastat-treated males: −7.2 [−14.2, 27.9]
    ERT-treated males: −1.17 [−15.6, 15.7]
  • Independent of treatment, yearly changes in cardiac parameters including LVMI in females and males were stable, pointing at disease stabilization
Van der Veen 2022 [33]0.5 mg/kg agalsidase beta: 3
1.0 mg/kg agalsidase beta: 4
Untreated: 23
Median [range]:
10.4 [9.5, 10.7] years
MRI and Echocardiography
  • Compared outcomes in young male patients (median [range]: treated, 24 [14, 26] years; untreated, 22 [13, 27] years) after 10 years of treatment with ERT with those of a group of untreated patients with FD with the same phenotype (classic) and of comparable age
  • 6/7 patients switched to full dose (1 mg/kg EOW) after a median treatment duration of 9 years (range: 8.3, 10)
  • LVM median [range] g/m2 at follow-up (baseline values NR)
    Based on echocardiography
    -
    Treated: 80 [67, 84]
    -
    Untreated: 94 [59, 149]
    -
    p = 0.02
    Based on MRI (including papillary muscles)
    -
    Treated: 53 [46, 59]
    -
    Untreated: 68 [53, 99]
    -
    p = 0.02
  • Analyses were repeated after excluding patients with renal involvement; this did not change the outcome
  • Mass measured by echocardiography and MRI correlated well (ϱ = 0.72, p = 0.002)
Vedder 2007 [28]Agalsidase alfa: 18
Agalsidase beta: 16
24 months
Echocardiography
  • Median LVM (SD) [range], g
    12 months: agalsidase beta, 296 (76) [169, 401]; agalsidase alfa, 244 (89) [157, 424]
    24 months: agalsidase beta, 308 (90) [196, 471]; agalsidase alfa, 294 (87) [196, 502]
  • No significant differences between groups
* Congress abstract. B.i.d., twice a day. CI, confidence interval. EOW, every other week. ERT, enzyme replacement therapy. ESRD, end-stage renal disease. FD, Fabry disease. FOS, Fabry Outcome Survey. IQR, interquartile range. LS, least-squares. LSM, least-squares mean. LVH, left ventricular hypertrophy. LVM, left ventricular mass. LVMI, left ventricular mass index. max, maximum. MRI, magnetic resonance imaging. NA, not applicable. NR, not reported. NS, non-significant. OLE, open-label extension. OR, odds ratio. q2w, every 2 weeks. q4w, every 4 weeks. RCT, randomized controlled trial. RRT, renal replacement therapy. SD, standard deviation. SE, standard error. SEM, standard error of the mean.

3.3. Cerebrovascular Outcomes

Cerebrovascular disease is a common manifestation of FD [1]. Overall, 24 studies assessing cerebrovascular outcomes were identified, with publications describing single-arm studies of agalsidase alfa (n = 6) [21,48,51,60,171,172], agalsidase beta (n = 4) [30,71,80,148], mixed or non-specified ERT (n = 12) [31,32,34,35,36,76,88,89,90,173,174,175], and other treatments (n = 2, both on migalastat) [101,176]. In total, eight publications reported the results of comparator studies [33,37,108,126,163,177,178,179] and one publication described a switch study [131]. An overview of cerebrovascular data from agalsidase alfa and agalsidase beta single-arm studies and the comparator studies is presented in Table 3. Key results from the other types of studies are provided in Supplementary Table S8.
The six agalsidase alfa single-arm studies of cerebrovascular outcomes had follow-up periods ranging from 6 months to 4 years (Table 3A). The largest of these was a long-term study in 677 patients from the FOS registry, of whom 85.4% had no cerebrovascular events over 3 years of treatment with agalsidase alfa [21]. Differences were noted in the frequency of cerebrovascular events between males and females, although these were inconsistent between studies [21,48]. Some smaller studies reported the development of WMLs in patients during treatment, possibly related to aging [51,60,171]. In the four agalsidase beta single-arm studies reporting on cerebrovascular outcomes, follow-up periods ranged from 1 to 20 years (Table 3A). Two studies reported no cerebrovascular events after at least 12 or 24 months of follow-up (n = 39 and 9, respectively) [30,148], although the development of new lesions in two asymptomatic male patients was noted in one of these studies [30]. In another study of Fabry Registry data, 10% of patients had a stroke or TIA over a median treatment duration of 6 years (n = 40) [80].
Single-arm studies of the effects of mixed or non-specified ERT on cerebrovascular outcomes in adults had median follow-up periods of 3.5 to 9 years, and all found that some patients receiving ERT developed cerebrovascular events (such as stroke or TIA) and/or WMLs (Supplementary Table S8). Two congress publications reported cerebrovascular outcomes in patients receiving migalastat in single-arm studies (one meta-analysis and one observational study) [101,176]. The meta-analysis of data from phase 2 and 3 clinical trials (n = 114) found that 7% of patients had cerebrovascular events over a median treatment duration of 4.4 years, with 2% having a first cerebrovascular event during treatment [176]. The observational study (n = 97) reported an incidence of 13.2 cerebrovascular events per 1000 patient-years over a median follow-up of 5.1 years [101].
Overall, studies reporting the efficacy of ERT versus placebo or other comparators in relation to cerebrovascular outcomes did not show a significant effect of treatment (Table 3B). One observational study (n = 112) investigated cerebrovascular outcomes in patients who either remained on agalsidase beta throughout the whole study, switched from agalsidase beta to alfa or switched from agalsidase beta to alfa and switched back to beta [131] (Supplementary Table S8). Over 24 months, the relative risk for stroke/TIA was lower in the agalsidase beta and switch groups than in the re-switch group [131].
Table 3. Overview of cerebrovascular data from (A) single-arm agalsidase alfa or agalsidase beta studies and (B) comparator studies.
Table 3. Overview of cerebrovascular data from (A) single-arm agalsidase alfa or agalsidase beta studies and (B) comparator studies.
(A)
Author, YearNTreatment DurationKey Results
Agalsidase alfa single-arm studies
Beck 2018 [21]
FOS
Total: 677
Female:
317 (47%)
Male:
360 (53%)
Median [IQR]:
Overall:
3.0 (1.1, 6.7) years;
Female:
2.9 (1.2, 6.3) years
Male:
3.2 (1.0, 7.0) years
  • Observational study from FOS
  • 578 patients (85.4%) (258 female [81.4%] and 320 male [88.9%]) did not experience a CBV event (stroke/TIA/PRIND) during follow-up
  • The Kaplan–Meier point estimate (95% CI) for median age in years at first CBV event was 70.9 (68.1, NE; due to the small number of events) in female patients and NE (64.9, NE) in male patients
Hughes 2011 [48]
FOS
Total: 250
Female:
78 (31.2%)
Male:
172 (68.8%)
4 years
  • Observational study using data from FOS
  • Retrospective chart review to determine whether women respond to ERT in a similar way to men
  • Data on CBV events (stroke/TIA/PRIND) were available for 21 women (27%)
  • 4 women (19%) experienced a CBV event at baseline
  • 1 (25%) of these 4 patients was reported to have also had an event between 42 and 54 months after the start of ERT
  • Of the 17 women (81%) who did not experience a CBV event at baseline,
    1 (6%) experienced an event between 42 and 54 months after the start of ERT
  • Data on CBV events were available for 23 men (13%)
  • Of the 7 men (30%) in whom a CBV event was reported at baseline, 4 (57%) were noted to have had a subsequent event at the 4-year time point
  • Among the 16 male patients (70%) who were not reported to have had an event at baseline, there were 2 (13%) new reports of a CBV event
Jardim 2006 [171]824 months
  • Prospective observational open-label study observing central nervous system manifestations during ERT
  • At baseline, 4 patients (50%) showed a widespread pattern of deep WML
  • In this group, WML showed a trend related to age and to the presence of hypoacusia
  • After 12 months of ERT, MRI remained normal in 4 patients (50%), showed the same WML in 2 other patients (25%) and showed worsening in
    2 patients (25%) who were the eldest
  • Of the 6 (75%) who completed 24 months on ERT, MRI was stable in 3 (50%); it was normal in 2 patients and showed the same lesions in the other patient. WML worsened in 1 patient (16.7%), fluctuated in 1 patient (16.7%) and disappeared in 1 patient (16.7%), who was the youngest
  • These variations in WML numbers did not correlate with residual enzymatic activity
  • In 2 years, 4 lesions disappeared and 8 appeared
  • Number of WML by patient (age in years at baseline), at baseline/12 months/24 months
    Patient 1 (46): 6/9/10
    Patient 2 (36): 6/8/8
    Patient 3 (31): 1/1/1
    Patient 4 (27): 0/0/0
    Patient 5 (27): 0/0
    Patient 6 (24): 2/2/0
    Patient 7 (24): 0/0
    Patient 8 (NR): 0/0/0
Lee 2017 [172]
FOS
Total: 37
IVS4: 25
Classic FD: 12
NR
  • Retrospective analysis comparing central nervous system manifestations in Taiwanese patients with the IVS4 mutation or classic FD mutations
  • Infarcts were observed in similar proportions of patients with IVS4 and classic FD (32.0% and 33.3%, respectively), although there were differences in the site of occurrence
  • Anterior circulation stroke and posterior circulation stroke alone each occurred in 8.0% of patients with IVS4
  • Anterior and posterior circulation stroke occurred in combination in 16.0% of patients with IVS4
  • Posterior circulation stroke alone was not observed in patients with classic FD
  • Anterior circulation stroke alone and anterior and posterior circulation stroke combined each occurred in 16.7% of classic patients with FD
  • There was a high prevalence of non-specific signs and symptoms or silent infarcts in both groups (62.5% in IVS4 and 50.0% in classic FD)
  • The pulvinar sign was observed in a greater proportion of patients with classic FD (50.0%) than IVS4 (32.0%)
  • Hemorrhage was noted in MRIs from 16.7% of patients with classic FD but not in patients with IVS4 (p = 0.0991)
  • Median (range) basilary artery diameter (mm) was 2.7 (1.4, 4.0) in patients with IVS4 and 3.2 (2.3, 4.7) in patients with classic FD (p = 0.0293)
Ries 2006 [51]24 pediatric patients24 weeks
  • Open-label study at 3 tertiary care centers in pediatric patients
  • One CBV accident occurred and was classed as a serious adverse event, considered unrelated to treatment
  • One 16-year-old patient had repeated small-vessel strokes, commencing at 14 years of age, which continued after >18 months on ERT
  • The patient had an intensive work-up that did not uncover a prothrombotic disorder other than FD. However, an unrecognized susceptibility factor for strokes (in addition to FD) was not excluded owing to the uncommonness of recurrent small-vessel strokes at that age
Thofehrn 2009 [60]936 months
  • Single-center, open-label study in Brazil
  • One patient had a transient CBV ischemic episode at 8 months of ERT and received aspirin and ticlopidine
Agalsidase beta single-arm studies
Cabrera 2017 [30]39 (switching owing to treatment shortage was allowed)≥12 months
  • Long-term prospective and historical analysis of cohort of patients from
    Argentina
  • Observational cohort study analyzing the effectiveness of long-term follow-up of ERT on renal, cardiac and CBV parameters
  • No patient had a stroke at baseline or during the follow-up
  • Two male patients presented progression in the number of new MRI lesions, all were asymptomatic
Dutra-Clarke 2021 [71]Total: 26
Adults
Female: 13 (50%)
Male: 11 (42%)
Children
Male: 2 (8%)
2–20 years
  • Retrospective cohort study in the US
  • WMLs were present in 4 adult participants (25%) who had brain imaging (3/10 female patients [30%], 1/6 male patients [17%])
  • One male patient in this cohort had a history of two CBV accidents, one TIA at 52 years of age and one stroke at 60 years of age
  • Another male patient and one female patient in this cohort had a history of TIAs while on ERT; they were not taking low dose aspirin at the time
  • Adult male patients had higher frequencies of clinical manifestations than adult female patients in all categories, with the exception of neuropathic pain, WMLs and corneal verticillate
Pisani 2005 [148]924 months
  • Non-randomized, open-label, prospective study evaluating the impact of 2 years of ERT on the clinical expression of FD in patients undergoing maintenance dialysis, and to investigate changes in symptoms and the echocardiographic evolution of FD cardiomyopathy in the context of RRT
  • 2 patients (22%) had CBV symptoms (TIA and stroke) at baseline (before starting ERT)
  • During the entire follow-up period, no patient experienced CBV events, and antihypertensive therapy had to be intensified in only 2 patients
Weidemann 2013 [80]40Median [IQR]:
6 (5.1, 7.2) years
  • Prospective observational study in patients with advanced FD at a single center in Germany
  • 4 patients (10%) developed a stroke during follow-up at ages 41, 46, 54, and 60 years
  • Only 1 of these patients experienced a TIA before the onset of stroke
  • 3 patients (8%) developed new TIA
(B)
Author, YearNTreatment DurationKey Results
Comparator studies
Banikazemi 2007 [177]Agalsidase beta: 51
Placebo: 31
Mean (SD):
18.4 (8.8) months
  • Randomized, double-blind, multi-center placebo-controlled trial
  • There were no stroke or TIA events in the agalsidase beta group compared with 2 strokes in the placebo group (OR: 0 [95% CI: 0, 3.2]; p = 0.14)
Fellgiebel 2014 [178]Agalsidase beta: 25
Placebo: 16
Mean [range]:
27 [12, 33] months
  • Post hoc analysis of data from a phase 4 placebo-controlled study
  • The effect of agalsidase beta on WML progression and CBV events were assessed longitudinally
  • At baseline, MRIs showed signs of brain infarction in 20% (5 of 25) of patients in the ERT group and 19% (3/16) of patients in the placebo group
  • At follow-up, MRIs showed that 13% (2/16) of patients in the placebo group and 4% (1 of 25) of patients in the ERT group had
  • developed new infarcts
  • WML burden was present in 63% of patients at baseline and correlated with LVH and previous or recent strokes
  • Mean normalized WML diameter (SD) at baseline, follow-up, follow-up minus baseline:
    Agalsidase beta (n = 25): 3.5 (5.2), 4.2 (5.6), 0.7 (1.9)
    Placebo (n = 16): 2.8 (3.0), 3.9 (4.3), 1.0 (2.1)
  • Overall, there were no significant differences in WML burden between the ERT and placebo groups at baseline or at follow-up
  • Patients > 50 years had a significantly higher WML burden at follow-up and significantly higher increases in WML burden over time compared with patients aged ≤ 50 years
  • In a subgroup analysis of patients ≤ 50 years of age, the proportion of patients with a stable or decreased WML load was significantly higher in the agalsidase beta group, (8/18 [44%]) than in the placebo group (4/13 [31%]; p = 0.014)
Lenders 2015 [179]ERT: 187
No ERT: 117
Mean [range]:
6.1 [1, 12]
  • Observational, retrospective study investigating the risk of thromboembolic events in Germany
  • The additional risk of thromboembolic events with the concurrence of FVL was assessed
  • Among all patients, 14.1% had experienced ≥ 1 stroke or TIA, while 3.3% experienced a deep vein thrombosis or pulmonary embolism
    Overall (n = 304): 89 thromboembolic events reported, 1.68 events per 100 person-years
    ERT (n = 8): 15 thromboembolic events, 1.94 events per 100 person-years
    No ERT (n = 15): 8 thromboembolic events, 3.73 events per 100 person-years
  • Patients with FD not receiving ERT have an ~2.8-fold increased risk of thromboembolic events compared with those receiving ERT
  • The occurrence of thromboembolic events was significantly increased for patients with FVL compared with those without FVL; HR: 5.45; 95% CI: 2.29, 12.99; p < 0.0001
Madsen 2017 [37]Agalsidase alfa or beta (switching allowed): 47
No ERT: 19
Median [range]:
ERT: 8 [0, 12] years;
No ERT: 6 [0, 13] years
  • Retrospective cohort study in a Danish cohort
  • At baseline, CBV events (stroke/TIA) occurred in 6 patients (13%) in the ERT group and 1 patient (5%) in the no ERT group
  • At follow-up, CBV events occurred in 7 patients (16%) in the ERT group and 0 patients (0%) in the no ERT group (p = 0.37)
Mignani 2008 [163]Dialysis: 17
Kidney transplant: 17
Mean (SD):
Dialysis: 45.1 (19.8) months;
Transplant: 48.4 (13.2) months
  • Cross-sectional survey study in Italian patients with FD and ESRD
  • During ERT, 9 acute cardiac and CBV events occurred in the dialysis group and 3 in the group of allograft recipients (p < 0.05); all were considered to unrelated to ERT
Skuban 2017 * [108]
FACETS
ATTRACT
FACETS
Migalastat or placebo: 67
OLE, migalastat: 60
FACETS: 6-month double-blind followed by open-label migalastat from 6 to 12 months (stage 2) plus an additional year
ATTRACT:
Migalastat or ERT: 53
18 months
  • Analysis of 2 randomized phase 3 clinical studies
  • ATTRACT: 1 CBV event (6%), a TIA, was reported in the ERT group
Van der Veen 2022 [33]Treated
Agalsidase beta: 3
Agalsidase alfa: 4
Untreated: 23
Median [range]: 10.4 [9.5, 10.7] years
  • Cross-sectional retrospective study comparing outcomes in patients with classic FD after 10 years of treatment with ERT to a group of untreated patients with the same phenotype of comparable age
  • WMLs were present in 1 treated patient (14%) and 6 untreated patients (26%) (p = 0.6)
  • In the untreated group, a lacunar infarction was described in one patient and microbleeds were found in another
Wilcox 2004 [126]Agalsidase beta: 29
Placebo: 29
30–36 months
  • Phase 3 randomized, placebo-controlled, double-blind trial
  • 5/58 (8.6%) patients reported CBV events (stroke or TIA) at the time of the report
  • 1 of these patients had an extensive past history of stroke; the other 4 patients, presumably, did not have a prior history of stroke
  • The lack of a placebo group makes interpretation of these data difficult
  • To note, the phase 3 extension study was not designed to assess the effect of ERT on clinical outcomes
  • Data on stroke and CV events were not collected as part of the study but rather as AEs
* Congress abstract. AE, adverse event. CBV, cerebrovascular. CI, confidence interval. CV, cardiovascular. ERT, enzyme replacement therapy. FD, Fabry disease. FOS, Fabry Outcome Survey. FVL, factor V Leiden. HR, hazard ratio. IQR, interquartile range. MRI, magnetic resonance imaging. NE, not estimable. NR, not reported. OLE, open-label extension. OR, odds ratio. PRIND, prolonged reversible ischemic neurologic deficit. RRT, renal replacement therapy. SD, standard deviation. TIA, transient ischemic attack. WML, white matter lesion.

3.4. Disease Severity

The MSSI was developed to measure the severity of general, renal, cardiovascular, and neurological signs and symptoms of FD and to monitor the clinical course of disease response to treatment [180]. The MSSI ranges from 0 to 76, with higher scores indicating greater disease severity. Scores are categorized as mild (<20 points), moderate (20–40 points), and severe (>40 points) [180]. Overall, 15 studies were identified that explored the impact of FD treatment on MSSI scores: four single-arm studies of agalsidase alfa [63,180,181,182], one with mixed ERT [28], two with other treatments (migalastat and pegunigalsidase alfa) [107,159], four comparator studies [113,183,184,185,186], and four switch studies [131,133,136,137,187]. No single-arm studies with MSSI data were identified for agalsidase beta. An overview of MSSI data is presented in Table 4.
Three of the four single-arm studies on agalsidase alfa demonstrated significant improvements in MSSI scores after 1–4 years of treatment [63,180,181] (Table 4A). In the fourth study (n = 85), 77% of patients had no improvement in MSSI score, and 14% progressed to more severe disease after 1 year of home therapy [182]. A mixed ERT study including agalsidase alfa and agalsidase beta (n = 20) found that a severe MSSI score at baseline was associated with treatment failure in male patients after 2 years of follow-up [28]. In an observational study of migalastat (n = 16), no significant changes in MSSI scores were observed over 18 months of treatment [159]. An observational study of pegunigalsidase alfa (n = 19) showed improvements in each MSSI subscale after 12 months of treatment [107].
Four comparator studies reporting MSSI scores were identified, all of which compared patients receiving unspecified ERT with those not receiving ERT (Table 4B). Two studies found no significant differences in MSSI scores between males in either group, while females on ERT had significantly higher scores [183,185]. One study found a significant improvement in cardiovascular MSSI scores with ERT after 12 months of treatment, although the number of patients was not reported [184,186]. The final study, which used the adapted FOS-MSSI in patients who underwent a kidney transplantation, did not find any differences between the treatment groups [113]. Of the four switch studies, two showed stable MSSI scores after switching from agalsidase beta to agalsidase alfa for 12 or 24 months [137,187], one found similar stable scores after re-switching to agalsidase beta [131], and one showed similar increases in MSSI scores in patients regardless of switching [133].
Table 4. Overview of MSSI data in (A) single-arm studies and (B) comparator and switch studies.
Table 4. Overview of MSSI data in (A) single-arm studies and (B) comparator and switch studies.
(A)
Author, Year
Study Identifier
NTreatment DurationKey Results
Agalsidase alfa single-arm studies
Concolino 2017 [182]85Mean [range]:
1.9 years
[3 months,
4.5 years]
  • Multi-center observational study of Italian patients receiving home infusions
  • MSSI total score, mean [range]
    Diagnosis (n = 73): 16.1 [1, 55]
    Starting home therapy (n = 75): 18.6 [2, 56]
    Last follow-up (n = 74): 19.3 [2, 54]
Parini 2008 [181]30Median [range]:
2.9 [1.0, 6.2] years
  • Multi-center observational study in Italy
  • Change from baseline to follow-up in MSSI score, median [range]
    Total: −4 [−21, 6]; p < 0.0001
    General: −1 [−5, 3]; p = 0.0005
    Neurological: −1 [−13, 5]; p = 0.0056
    CV and renal MSSI scores did not change from baseline
Whybra 2004 [180]391 year
  • Single-center study of adults with FD in Germany
  • Decrease in MSSI score from baseline, median [range]
    Total: 9 [6, 12]; p < 0.001
    General: 2 [1, 3]; p < 0.001
    Neurological: 3 [2, 5]; p < 0.001
    CV: 2 [0, 4]; p < 0.001
    Renal: 0 [0, 2]
Whybra 2009 [63]404 years
  • Prospective single-center open-label study of female patients in Germany
  • MSSI total score decreased significantly after the first year of treatment and remained significantly reduced through 4 years
    Changes in the neurologic and CV subscores were responsible for most of the change in MSSI
Mixed ERT single-arm studies
Vedder 2007 [28]342 years
  • Randomized open-label study of Dutch and Norwegian patients
  • MSSI score, mean [range]
    Males with treatment failure (progression of cardiac, renal or cerebrovascular disease): 44 [17, 59]
    Males without treatment failure: 21 [12, 37]; p = 0.02 vs. with treatment failure
Migalastat single-arm studies
Camporeale 2023 [159]
MAIORA
1618 months
  • Prospective, observational single-center study to evaluate cardiac outcomes in Italian patients
  • MSSI (Median [range])
    Total score:
    baseline: 19.0 [16.0, 23.0]; follow-up: 18.5 [13.5, 24.5]; p = 0.31
    General score:
    baseline: 5.0 [3.0, 6.0]; follow-up: 5.0 [2.0, 6.0]; p = 0.31
    Neurological score:
    baseline: 5.0 [3.0, 7.0]; follow-up: 6.5 [3.5, 8.0]; p = 0.85
    CV score:
    baseline: 9.0 [0.0, 13.0]; follow-up: 9.5 [3.0, 13.0]; p = 1.00
    Renal score:
    baseline: 0.0 [0.0, 2.0]; follow-up: 0.0 [0.0, 0.0]; p = 1.00
Pegunigalsidase alfa single-arm studies
Schiffmann
2019 [107] NCT01678898, NCT01769001
1912 months
  • Improvements in each subscale of the MSSI were observed during the study
    Total MSSI score:
    baseline: 6.6, follow-up change from baseline: −1.9
    Neurological score:
    baseline: 7.8, follow-up change from baseline: −2.6
    CV score:
    baseline: 3.7, follow-up change from baseline: −0.8
    Renal score:
    baseline: 2.5, follow-up change from baseline: −1.0
(B)
Author, year
Study Identifier
Treatment
Groups, n
Key Results
Comparator studies
Biegstraaten 2010 [183]ERT: 30
No ERT: 18
  • Cohort from the Netherlands
  • MSSI total/general/neurological/CV/renal, median [range]
    ERT male (n = 13)
    -
    27 [10, 47]/5 [2, 11]/6 [3, 13]/9 [1, 15]/0 [0, 12]
    Non-ERT male (n = 2)
    -
    11 [5, 17]; p = 0.23 vs. ERT male/3.5 [2, 5]; p = 0.31 vs. ERT male/0.5 [0, 1]; p = 0.02 vs. ERT male/7 [3, 11]; p = 0.69 vs. ERT male/0; p = 0.38 vs. ERT male
    ERT female (n = 17)
    -
    23 [3, 35]/5 [1, 8]/4 [1, 12]/9 [0, 15]/4 [0, 12]
    Non-ERT female (n = 16)
    -
    7 [1, 18]; p = 0.00 vs. ERT female/2 [0, 5]; p = 0.00 vs. ERT female/1.5 [0, 7]; p = 0.04 vs. ERT female/1.5 [0, 11]; p = 0.07 vs. ERT female/0; p = 0.00 vs. ERT female
Chen 2016 [184], Chen 2017 [186]ERT: 25
  • MSSI CV score from baseline to 12 months, mean (SD)
  • ERT baseline: 13.8 (2.8)
    ERT follow-up: 12.4 (2.3); p < 0.05 vs. baseline
    Cybulla 2008 [113]
    FOS
    Kidney transplant + ERT: 20
    Kidney transplant + no ERT: 7
    • FOS-MSSI score, mean (SD) at baseline
      ERT: 34.7 (9.2)
      No ERT: 31.6 (6.3)
    Rosa 2021 [185]ERT: 22
    No ERT: NR
    • MSSI score, mean (SD) at baseline
      ERT men: 29.0 (11.1)
      No ERT men: 26.7 (14.9); p = 0.78 vs. ERT men
      ERT women: 25.8 (8.2)
      No ERT women: 10.3 (7.1); p = 0.0003 vs. ERT women
    Switch studies
    Kramer 2018 [131]Regular dose agalsidase beta: 37
    Switch group to agalsidase alfa: 38
    Re-switch group to agalsidase beta after 12 months agalsidase alfa: 37
    • MSSI score, after 12 months of treatment/24 months of treatment, mean (SD)
      Regular dose: 25 (13)/27 (12)
      Switch: 23 (12)/25 (12)
      Re-switch: 21 (10)/22 (11)
      p < 0.05 switch v re-switch after 12 months
    Lenders 2021 [133]Agalsidase beta regular dose: 17
    Switch: 22 (patients treated with agalsidase beta for
    ≥12 months, then dose-reduced and subsequently switched to agalsidase alfa)
    Re-switch: 39 (patients treated with agalsidase beta for
    ≥12 months, then dose-reduced or switched to agalsidase alfa for
    ≥24 months and then re-switched to agalsidase beta)
    • All groups showed a significant increase in MSSI scores over the follow-up period of ~80 months
      Re-switch group: MSSI increase: 1.0 (95% CI: 0.7, 1.4) per year (p < 0.001)
      Switch group: MSSI increase: 0.8 (95% CI: 0.2, 1.5) per year(p = 0.0189)
      Regular agalsidase-beta group: MSSI increase: 1.1 (95% CI: 0.7, 1.4) per year (p < 0.001)
    • Comparison between groups: no significant differences in yearly MSSI increase between the three groups (p = 0.7483)
    Ripeau 2017 [187]Agalsidase beta switch to agalsidase alfa: 33
    • MSSI score, mean (SD)
      Baseline: 22.1 (1.4)
      24 months: 22.5 (1.4); p = NS vs. baseline
    Tsuboi 2012 [136], Tsuboi 2014 [137]Agalsidase beta switch to agalsidase alfa: 11
    • MSSI score general/neurological/CV/renal/total, mean
      Baseline of switch: 4.09/5.18/8.73/4.73/22.73
      After 12 months: 3.91/5.00/8.73/4.73/22.36
    CV, cardiovascular. ERT, enzyme replacement therapy. FD, Fabry disease. FOS, Fabry Outcome Survey. IQR, interquartile range. MSSI, Mainz Severity Score Index. NR, not reported. NS, non-significant. SD, standard deviation.

    4. Discussion

    This SLR identified a total of 247 publications reporting findings from 231 studies that reported data on renal, cardiovascular, cerebrovascular, and disease severity (MSSI) outcomes with FD treatments. The majority of these were real-world studies involving adults. As expected, the approved treatments for FD were well represented in the literature, with 187 studies evaluating ERT with agalsidase alfa and/or agalsidase beta, 30 studies examining migalastat, and 11 studies investigating pegunigalsidase alfa. The treatments for which the longest follow-up data were available were agalsidase alfa (up to 20 years [40,43]) and agalsidase beta (up to 10 years [82]). In general, FD treatments stabilized or slowed kidney function decline and cardiac structural changes. However, in the absence of a randomized control group, as was the case for many of the studies, and owing to the variable presentation and slow progression of FD, such findings should be interpreted with caution. There was substantial heterogeneity in the disease severity and clinical characteristics of patients at baseline, both within and between studies, as well as differences in study designs with regard to the length of follow-up, sex balance, age groups, treatment duration, and treatment doses. As a result, cross-study conclusions about the effectiveness of different therapies cannot be accurately made. This is aligned with SLRs by Orsborne et al. (2023) [188], Besekar et al. (2023) [189], and Riccio et al. (2023) [190,191].
    FD can significantly impair kidney function, leading to reduced GFR and increased proteinuria [1]. The results of this SLR suggest that ERT with agalsidase alfa or agalsidase beta has renoprotective effects and stabilizes GFR over time. Migalastat also demonstrated stability in renal function in many studies. Cardiac manifestations of FD, including LVH, also contribute significantly to morbidity and mortality [1]. ERT with agalsidase alfa or agalsidase beta or oral chaperone therapy with migalastat stabilized or reduced LVMI, indicating beneficial effects on cardiac structure. A study of 2171 patients from FOS treated with agalsidase alfa for up to 20.8 years (published in April 2025) also demonstrated improved renal and cardiac outcomes compared with external untreated cohorts [192]. Renal outcomes improved regardless of sex or baseline proteinuria, and LVMI stabilized regardless of sex or baseline LVH status. Agalsidase alfa also delayed composite morbidity events and mortality compared with untreated external cohorts [192].
    Evidence from studies directly comparing agalsidase alfa and agalsidase beta (limited to two cohort studies and one randomized, controlled, open-label trial) suggests they have similar efficacy with respect to renal and cardiac outcomes. The importance of early treatment initiation was demonstrated, with early initiation of ERT in childhood or young adulthood, before established organ involvement, associated with improved preservation of GFR outcomes and LVMI. In subgroup analyses of ERT studies, patients with impaired kidney function/high renal involvement at baseline experienced a greater decline in eGFR than those with preserved kidney function/low renal involvement at baseline.
    Patients with FD are also at increased risk of cerebrovascular events, such as strokes [1]. Studies so far have provided little evidence of a benefit with ERT or migalastat treatment on cerebrovascular outcomes. Comparisons between different ERTs, which do not cross the blood–brain barrier, and migalastat, which may be able to cross the blood–brain barrier [193], require further investigation to establish the optimal treatment strategy for cerebrovascular risk reduction. However, the improvements in cardiovascular function observed with ERT and migalastat treatment may contribute to better cerebrovascular outcomes. After the completion of the literature search for this SLR (17 June 2024), the incidence of stroke was reported in a publication from the Fabry Registry [194]. In agalsidase beta-treated patients, the stroke incidence was 5.55 per 1000 person-years compared with 11.18 per 1000 person-years in matched untreated patients and agalsidase beta-treated patients had a significantly lower risk of stroke compared with untreated patients (HR: 0.36; 95% CI: 0.23, 0.56) [194].
    Compared with other outcomes of interest, there was a paucity of research into the effects of FD treatments on MSSI, which incorporates multiple clinical parameters to assess and monitor FD severity as a multisystem disease and helps to stratify patients by clinical phenotype [180]. Agalsidase alfa showed improvements in MSSI scores in several studies (particularly in the general, neurological, and cardiovascular subscores), whereas the effects of agalsidase beta on MSSI scores remain less explored. Studies on the impact of switching treatments showed stability in MSSI scores, suggesting comparable efficacy between agalsidase alfa and beta. Although not evaluated in this SLR, ERT has been shown to improve neuropathic pain and quality of life in patients with FD [195].
    Following the completion of the literature search for this SLR (17 June 2024), nine further clinical studies reporting renal and cardiac outcomes in FD have been published. This comprised two studies of agalsidase alfa, two studies of agalsidase beta, four studies of migalastat, and one study of pegunigalsidase alfa [196,197,198,199,200,201,202,203,204]. Overall, these publications support the efficacy of ERT and migalastat (in patients with amenable GLA variants) on improving renal and cardiac function and reducing the risk of severe clinical events.
    The slow and variable progression of FD mean that careful baseline phenotyping and long-term follow-up is required to demonstrate the clinical benefit–risk profile of treatments and patient adherence with them. Results from long-term studies are awaited for the more recent pegunigalsidase alfa, gene therapy, and substrate reduction therapies. Regarding cardiac outcomes, the major limitation of most of the clinical trials is the limited follow-up time and the inadequacy of surrogate biomarkers for cardiomyopathy [167]. While LVM and LVMI measure the hypertrophic aspect of cardiac deterioration, they do not reflect the risk of arrhythmias and diastolic heart failure, which are the prevalent complications in patients with FD [167]. There were also limited data on the use of additional supportive medications in patients with FD, such as renin–angiotensin blockade, anti-platelet therapies etc. Patients with FD often receive multiple treatments, such as those to manage primary and secondary cardiovascular risks. Use of concomitant medicines are also likely to affect treatment outcomes [15]. Therefore, future studies should address the potential effect of concomitant medications on the efficacy of FD treatments.
    Published congress abstracts were included in this SLR to capture all available published data on the outcomes of interest. Unlike journal articles, however, congress publications are typically not peer reviewed and often contain limited study details. Therefore, the results of such studies should be considered with caution. Different studies were identified in this SLR compared with previous SLR reports [188,189,190], owing to differences in methodology and focus. Laboratory measurements, such as serum levels of globotriaosylsphingosine (lyso-Gb3) or the presence of antidrug antibodies, have been suggested to relate to the effectiveness of FD treatments; however, they do not translate directly to clinical outcomes and were therefore not evaluated in this SLR. Although safety outcomes were reported in various identified publications, these outcomes have not been covered here owing to the heterogenous nature of safety reporting in the clinical studies.
    A major limitation of this SLR is the small number and heterogeneous phenotypes of patients enrolled in many of the identified clinical trials and observational studies. Although this issue is common in studies of rare diseases, such limited sample sizes may lead to difficulty in generalizing results for larger populations. In addition, follow-up times varied substantially between studies. Long-term follow-up studies with larger patient cohorts, stratified according to clinical phenotype, will be essential for the accurate assessment of the sustained benefits and potential side effects of FD treatments over extended periods. The variability of study designs and the small treatment effects also meant that it was not possible to directly compare different subgroups and studies, so the differential effects for certain patient groups could not be ascertained. Furthermore, studies comparing agalsidase alfa and agalsidase beta had significant limitations. Specifically, studies with a retrospective design can introduce significant biases related to patient selection and data collection, while open-label studies may suffer from biases in data analysis and interpretation. Future studies should address the effects of different FD treatments in different age groups on specific outcomes, with methodology that is reproducible enough to produce robust data for meta-analysis.
    In conclusion, after more than 20 years of experience with ERT, data indicate that agalsidase alfa and agalsidase beta demonstrate similar efficacy in stabilizing renal and cardiac outcomes in patients with FD. Early initiation of ERT in childhood or young adulthood before established organ involvement is critical for optimizing treatment outcomes. Treatment with migalastat has also been associated with the stabilization of renal and cardiac outcomes in patients with amenable GLA variants. Large cohort studies of current and emerging treatments will require long-term follow-up periods. In addition, better stratification according to clinical phenotype and prognosis will be crucial for addressing evidence gaps and providing clinicians and policymakers with comprehensive and reliable data to offer informed treatment options and share treatment decisions with their patients.

    Supplementary Materials

    The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm14145131/s1, Figure S1: Study locations; Table S1: EMBASE, MEDLINE and Cochrane search strategies; Table S2: Eligibility search criteria; Table S3: Risk of bias assessment for included studies; Table S4: Overview of GFR data from (A) mixed or non-specified ERT single-arm studies and single-arm studies for other treatments and (B) switch studies; Table S5: Overview of proteinuria data from (A) single-arm studies and (B) comparator and switch studies; Table S6: Overview of LVMI data from (A) mixed or non-specified ERT single-arm studies and single-arm studies for other treatments and (B) switch studies; Table S7: Overview of data for other structural cardiovascular parameters from (A) single-arm studies, (B) comparator and (C) switch studies; Table S8: Overview of cerebrovascular data from (A) mixed or non-specified ERT single-arm studies and single-arm studies for other treatments and (B) switch studies.

    Author Contributions

    Conception of the analysis: F.C.; Development of the protocol: F.C., O.E. and C.S.; Performing the systematic literature review: F.C., O.E. and C.S.; Interpretation of the results: A.J., E.M.-H., F.C., O.E., O.A., D.H., G.P.-M., R.G. and S.F.; Contribution to drafting the manuscript or to revising it critically for important intellectual content: C.S., A.J., E.M.-H., F.C., O.E., O.A., D.H., G.P.-M., R.G. and S.F. All authors have read and agreed to the published version of the manuscript.

    Funding

    The systematic literature review was funded by Takeda Development Center Americas, Inc.

    Data Availability Statement

    The data included in this report are from the published literature.

    Acknowledgments

    Under the direction of the authors, medical writing support was provided by Sarah Graham of PharmaGenesis, London, UK, and funded by Takeda Development Center Americas, Inc. in accordance with Good Publication Practice (GPP 2022) guidelines (www.ismpp.org/gpp-2022 [accessed on 14 October 2024]).

    Conflicts of Interest

    Ana Jovanovic has received honoraria for speaking and advisory boards from Amicus, Chiesi, Sanofi, and Takeda and research grants from Amicus and Takeda. Eve Miller-Hodges has received consultancy fees from Takeda and Amicus and speakers’ fees from Sanofi and Amicus. Felicia Castriota is an employee of Takeda Development Center Americas, Inc. and owns Takeda stock. Obaro Evuarherhe is an employee of Oxford PharmaGenesis Ltd. Olulade Ayodele is an employee of Takeda Development Center Americas, Inc. and owns Takeda stock. Derralynn Hughes has received honoraria for speaking and advisory boards from Amicus, Chiesi, Freeline, Idorsia, Sanofi, and Takeda. Guillem Pintos-Morell has received honoraria from Alexion, Amicus, Chiesi, Immedica, Lucane, Sanofi, Takeda, and Vitaflo and unrestricted grants from Sanofi and Takeda to the Vall d’Hebron Research Foundation for funding research on rare diseases. Roberto Giugliani has received consulting fees, honoraria for speaking and advisory boards, and/or research funding from Alexion, Alnylam Pharmaceuticals, Amicus Therapeutics, Astellas Pharma, Azafaros, BioMarin Pharmaceutical, Chiesi, JCR Pharmaceutical, Novartis, Paradigm BioPharmaceuticals, Passage Bio, Praxis Precision Medicines, PTC Therapeutics, Regenxbio, Sanofi, Takeda, and Ultragenyx. He is a member and the current chair of the Fabry Outcome Survey Steering Committee. Sandro Feriozzi has received honoraria for speaking and advisory boards from Amicus, Chiesi, Otsuka, Sanofi, and Takeda. Csaba Siffel is an employee of Takeda Development Center Americas, Inc. and owns Takeda stock.

    References

    1. Germain, D.P. Fabry disease. Orphanet J. Rare Dis. 2010, 5, 30–79. [Google Scholar] [CrossRef] [PubMed]
    2. Biegstraaten, M.; Arngrímsson, R.; Barbey, F.; Boks, L.; Cecchi, F.; Deegan, P.B.; Feldt-Rasmussen, U.; Geberhiwot, T.; Germain, D.P.; Hendriksz, C.; et al. Recommendations for initiation and cessation of enzyme replacement therapy in patients with Fabry disease: The European Fabry Working Group consensus document. Orphanet J. Rare Dis. 2015, 10, 36. [Google Scholar] [CrossRef] [PubMed]
    3. MacDermot, K.D.; Holmes, A.; Miners, A.H. Anderson-Fabry disease: Clinical manifestations and impact of disease in a cohort of 98 hemizygous males. J. Med. Genet. 2001, 38, 750–760. [Google Scholar] [CrossRef] [PubMed]
    4. MacDermot, K.D.; Holmes, A.; Miners, A.H. Anderson-Fabry disease: Clinical manifestations and impact of disease in a cohort of 60 obligate carrier females. J. Med. Genet. 2001, 38, 769–775. [Google Scholar] [CrossRef] [PubMed]
    5. Germain, D.P.; Altarescu, G.; Barriales-Villa, R.; Mignani, R.; Pawlaczyk, K.; Pieruzzi, F.; Terryn, W.; Vujkovac, B.; Ortiz, A. An expert consensus on practical clinical recommendations and guidance for patients with classic Fabry disease. Mol. Genet. Metab. 2022, 137, 49–61. [Google Scholar] [CrossRef] [PubMed]
    6. Mehta, A.; Hughes, D.A. Fabry disease. In GeneReviews® [Internet]; Adam, M.P., Feldman, J., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Amemiya, A., Eds.; Updated 11 April 2024; University of Washington: Seattle, WA, USA, 2002. Available online: https://www.ncbi.nlm.nih.gov/books/NBK1292/pdf/Bookshelf_NBK1292.pdf (accessed on 14 October 2024).
    7. Arends, M.; Wanner, C.; Hughes, D.; Mehta, A.; Oder, D.; Watkinson, O.T.; Elliott, P.M.; Linthorst, G.E.; Wijburg, F.A.; Biegstraaten, M.; et al. Characterization of classical and nonclassical Fabry disease: A multicenter study. J. Am. Soc. Nephrol. 2017, 28, 1631–1641. [Google Scholar] [CrossRef] [PubMed]
    8. European Medicines Agency. Replagal. Summary of Product Characteristics 2022. Available online: https://www.ema.europa.eu/en/documents/product-information/replagal-epar-product-information_en.pdf (accessed on 14 October 2024).
    9. European Medicines Agency. Fabrazyme. Summary of Product Characteristics 2024. Available online: https://www.ema.europa.eu/en/documents/product-information/fabrazyme-epar-product-information_en.pdf (accessed on 14 October 2024).
    10. Food and Drug Administration. Fabrazyme. Prescribing Information 2024. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/103979s5313lbl.pdf (accessed on 14 October 2024).
    11. European Medicines Agency. Elfabrio. Summary of Product Characteristics 2023. Available online: https://www.ema.europa.eu/en/documents/product-information/elfabrio-epar-product-information_en.pdf (accessed on 14 October 2024).
    12. Food and Drug Administration. Elfabrio. Prescribing Information 2023. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/761161s000lbl.pdf (accessed on 14 October 2024).
    13. European Medicines Agency. Galafold. Summary of Product Characteristics 2023. Available online: https://www.ema.europa.eu/en/documents/product-information/galafold-epar-product-information_en.pdf (accessed on 14 October 2024).
    14. Food and Drug Administration. Galafold. Prescribing Information 2024. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/208623s008lbl.pdf (accessed on 14 October 2024).
    15. Lenders, M.; Brand, E. Fabry disease: The current treatment landscape. Drugs 2021, 81, 635–645. [Google Scholar] [CrossRef] [PubMed]
    16. Van der Veen, S.J.; Hollak, C.E.M.; van Kuilenburg, A.B.P.; Langeveld, M. Developments in the treatment of Fabry disease. J. Inherit. Metab. Dis. 2020, 43, 908–921. [Google Scholar] [CrossRef] [PubMed]
    17. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
    18. Bierer, G.; Balfe, D.; Wilcox, W.R.; Mosenifar, Z. Improvement in serial cardiopulmonary exercise testing following enzyme replacement therapy in Fabry disease. J. Inherit. Metab. Dis. 2006, 29, 572–579. [Google Scholar] [CrossRef] [PubMed]
    19. Bodensteiner, D.; Scott, C.R.; Sims, K.B.; Shepherd, G.M.; Cintron, R.D.; Germain, D.P. Successful reinstitution of agalsidase beta therapy in Fabry disease patients with previous IgE-antibody or skin-test reactivity to the recombinant enzyme. Genet. Med. 2008, 10, 353–358. [Google Scholar] [CrossRef] [PubMed]
    20. Hughes, D.; Linhart, A.; Gurevich, A.; Kalampoki, V.; Jazukeviciene, D.; Feriozzi, S. Prompt initiation of agalsidase alfa therapy is associated with improved cardiovascular and renal outcomes in the Fabry Outcome Survey (FOS). Mol. Genet. Metab. 2020, 129 (Suppl. S2), S77. [Google Scholar] [CrossRef]
    21. Beck, M.; Hughes, D.; Kampmann, C.; Pintos-Morell, G.; Ramaswami, U.; West, M.L.; Giugliani, R. Long-term outcomes with agalsidase alfa enzyme replacement therapy: Analysis using deconstructed composite events. Mol. Genet. Metab. Rep. 2018, 14, 31–35. [Google Scholar] [CrossRef] [PubMed]
    22. Feriozzi, S.; Linhart, A.; Ramaswami, U.; Kalampoki, V.; Gurevich, A.; Hughes, D. Effects of baseline left ventricular hypertrophy and decreased renal function on cardiovascular and renal outcomes in patients with Fabry disease treated with agalsidase alfa: A Fabry Outcome Survey study. Clin. Ther. 2020, 42, 2321–2330.e0. [Google Scholar] [CrossRef] [PubMed]
    23. Hopkin, R.J.; Cabrera, G.H.; Jefferies, J.L.; Yang, M.; Ponce, E.; Brand, E.; Feldt-Rasmussen, U.; Germain, D.P.; Guffon, N.; Jovanovic, A.; et al. Clinical outcomes among young patients with Fabry disease who initiated agalsidase beta treatment before 30 years of age: An analysis from the Fabry Registry. Mol. Genet. Metab. 2023, 138, 106967. [Google Scholar] [CrossRef] [PubMed]
    24. Hughes, D.; Linhart, A.; Gurevich, A.; Kalampoki, V.; Jazukeviciene, D.; Feriozzi, S. Prompt agalsidase alfa therapy initiation is associated with improved renal and cardiovascular outcomes in a Fabry Outcome Survey analysis. Drug Des. Dev. Ther. 2021, 15, 3561–3572. [Google Scholar] [CrossRef] [PubMed]
    25. Ortiz, A.; Abiose, A.; Bichet, D.G.; Cabrera, G.; Charrow, J.; Germain, D.P.; Hopkin, R.J.; Jovanovic, A.; Linhart, A.; Maruti, S.S.; et al. Time to treatment benefit for adult patients with Fabry disease receiving agalsidase beta: Data from the Fabry Registry. J. Med. Genet. 2016, 53, 495–502. [Google Scholar] [CrossRef] [PubMed]
    26. Parini, R.; Pintos-Morell, G.; Hennermann, J.B.; Hsu, T.-R.; Karabul, N.; Kalampoki, V.; Gurevich, A.; Ramaswami, U. Analysis of renal and cardiac outcomes in male participants in the Fabry Outcome Survey starting agalsidase alfa enzyme replacement therapy before and after 18 years of age. Drug Des. Dev. Ther. 2020, 14, 2149–2158. [Google Scholar] [CrossRef] [PubMed]
    27. Sirrs, S.M.; Bichet, D.G.; Iwanochko, R.M.; Khan, A.; Doucette, S.; Lemoine, K. Differential effects of agalsidase alfa and agalsidase beta in Fabry out-comes: 10 year outcomes from the Canadian Fabry Disease Initiative. J. Inherit. Metab. Dis. 2018, 41 (Suppl. S1), S188. [Google Scholar]
    28. Vedder, A.C.; Linthorst, G.E.; Houge, G.; Groener, J.E.; Ormel, E.E.; Bouma, B.J.; Aerts, J.M.; Hirth, A.; Hollak, C.E.; Schiffmann, R. Treatment of Fabry disease: Outcome of a comparative trial with agalsidase alfa or beta at a dose of 0.2 mg/kg. PLoS ONE 2007, 2, e598. [Google Scholar] [CrossRef] [PubMed]
    29. Arends, M.; Biegstraaten, M.; Wanner, C.; Sirrs, S.; Mehta, A.; Elliott, P.M.; Oder, D.; Watkinson, O.T.; Bichet, D.G.; Khan, A.; et al. Agalsidase alfa versus agalsidase beta for the treatment of Fabry disease: An international cohort study. J. Med. Genet. 2018, 55, 351–358. [Google Scholar] [CrossRef] [PubMed]
    30. Cabrera, G.; Politei, J.; Antongiovani, N.; Amartino, H. Effectiveness of enzyme replacement therapy in Fabry disease: Long term experience in Argentina. Mol. Genet. Metab. Rep. 2017, 11, 65–68. [Google Scholar] [CrossRef] [PubMed]
    31. Arends, M.; Biegstraaten, M.; Hughes, D.A.; Mehta, A.; Elliott, P.M.; Oder, D.; Watkinson, O.T.; Vaz, F.M.; van Kuilenburg, A.B.P.; Wanner, C.; et al. Retrospective study of long-term outcomes of enzyme replacement therapy in Fabry disease: Analysis of prognostic factors. PLoS ONE 2017, 12, e0182379. [Google Scholar] [CrossRef] [PubMed]
    32. Arends, M.; Wijburg, F.A.; Wanner, C.; Vaz, F.M.; van Kuilenburg, A.B.; Hughes, D.A.; Biegstraaten, M.; Mehta, A.; Hollak, C.E.; Langeveld, M. Favourable effect of early versus late start of enzyme replacement therapy on plasma globotriaosylsphingosine levels in men with classical Fabry disease. Mol. Genet. Metab. 2017, 121, 157–161. [Google Scholar] [CrossRef] [PubMed]
    33. Van der Veen, S.; Körver, S.; Hirsch, A.; Hollak, C.; Wijburg, F.; Brands, M.; Tøndel, C.; van Kuilenburg, A.; Langeveld, M. Early start of enzyme replacement therapy in pediatric male patients with classical Fabry disease is associated with attenuated disease progression. Mol. Genet. Metab. 2022, 135, 163–169. [Google Scholar] [CrossRef] [PubMed]
    34. Nowak, A.; Koch, G.; Huynh-Do, U.; Siegenthaler, M.; Marti, H.P.; Pfister, M. Disease progression modeling to evaluate the effects of enzyme replacement therapy on kidney function in adult patients with the classic phenotype of Fabry disease. Kidney Blood Press. Res. 2017, 42, 1–15. [Google Scholar] [CrossRef] [PubMed]
    35. Wyatt, K.; Henley, W.; Anderson, L.; Anderson, R.; Nikolaou, V.; Stein, K.; Klinger, L.; Hughes, D.; Waldek, S.; Lachmann, R.; et al. The effectiveness and cost-effectiveness of enzyme and substrate replacement therapies: A longitudinal cohort study of people with lysosomal storage disorders. Health Technol. Assess. 2012, 16, 1–543. [Google Scholar] [CrossRef] [PubMed]
    36. Anderson, L.J.; Wyatt, K.M.; Henley, W.; Nikolaou, V.; Waldek, S.; Hughes, D.A.; Pastores, G.M.; Logan, S. Long-term effectiveness of enzyme replacement therapy in Fabry disease: Results from the NCS-LSD cohort study. J. Inherit. Metab. Dis. 2014, 37, 969–978. [Google Scholar] [CrossRef] [PubMed]
    37. Madsen, C.V.; Bundgaard, H.; Rasmussen, Å.K.; Sørensen, S.S.; Petersen, J.H.; Køber, L.; Feldt-Rasmussen, U.; Petri, H. Echocardiographic and clinical findings in patients with Fabry disease during long-term enzyme replacement therapy: A nationwide Danish cohort study. Scand. Cardiovasc. J. 2017, 51, 207–216. [Google Scholar] [CrossRef] [PubMed]
    38. Silva, C.A.B.; Moura-Neto, J.A.; Reis, M.A.D.; Neto, O.M.V.; Barreto, F.C. Renal manifestations of Fabry disease: A narrative review. Can. J. Kidney Health Dis. 2021, 8, 2054358120985627. [Google Scholar] [CrossRef] [PubMed]
    39. Beck, M.; Ricci, R.; Widmer, U.; Dehout, F.; De Lorenzo, A.G.; Kampmann, C.; Linhart, A.; Sunder-Plassmann, G.; Houge, G.; Ramaswami, U.; et al. Fabry disease: Overall effects of agalsidase alfa treatment. Eur. J. Clin. Investig. 2004, 34, 838–844. [Google Scholar] [CrossRef] [PubMed]
    40. Giugliani, R.; Hughes, D.; Nicholls, K.; Niu, D.-M.; Reisin, R.C.; Botha, J.; Anagnostopoulou, C.; West, M.L. Long-term outcomes in patients with Fabry disease who were treated with agalsidase alfa for more than nineteen years: The Fabry Outcome Survey. Mol. Genet. Metab. 2023, 138, 107118. [Google Scholar] [CrossRef]
    41. Pintos-Morell, G.; Kampmann, C.; Botha, J.; Anagnostopoulou, C.; West, M.L.; Ramaswami, U. Early initiation of agalsidase alfa treatment improves clinical outcomes in male patients with classical Fabry disease: A Fabry Outcome Survey (FOS) analysis. Mol. Genet. Metab. 2023, 138, 107273. [Google Scholar] [CrossRef]
    42. Ramaswami, U.; Beck, M.; Hughes, D.; Kampmann, C.; Botha, J.; Pintos-Morell, G.; West, M.L.; Niu, D.M.; Nicholls, K.; Giugliani, R.; et al. Cardio-renal outcomes with long-term agalsidase alfa enzyme replacement therapy: A 10-year Fabry Outcome Survey (FOS) analysis. Drug Des. Dev. Ther. 2019, 13, 3705–3715. [Google Scholar] [CrossRef] [PubMed]
    43. Cybulla, M.; Nicholls, K.; Feriozzi, S.; Linhart, A.; Torras, J.; Vujkovac, B.; Botha, J.; Anagnostopoulou, C.; West, M.L. Renoprotective effect of agalsidase alfa: A long-term follow-up of patients with Fabry disease. J. Clin. Med. 2022, 11, 4810. [Google Scholar] [CrossRef] [PubMed]
    44. Feriozzi, S.; Schwarting, A.; Sunder-Plassmann, G.; West, M.; Cybulla, M.I.; International Fabry Outcome Survey Investigators. Agalsidase alfa slows the decline in renal function in patients with Fabry disease. Am. J. Nephrol. 2008, 29, 353–361. [Google Scholar] [CrossRef] [PubMed]
    45. Feriozzi, S.; Torras, J.; Cybulla, M.; Nicholls, K.; Sunder-Plassmann, G.; West, M.; FOS Investigators. The effectiveness of long-term agalsidase alfa therapy in the treatment of Fabry nephropathy. Clin. J. Am. Soc. Nephrol. 2012, 7, 60–69. [Google Scholar] [CrossRef] [PubMed]
    46. Goker-Alpan, O.; Longo, N.; McDonald, M.; Shankar, S.P.; Schiffmann, R.; Chang, P.; Shen, Y.; Pano, A. An open-label clinical trial of agalsidase alfa enzyme replacement therapy in children with Fabry disease who are naive to enzyme replacement therapy. Drug Des. Dev. Ther. 2016, 10, 1771–1781. [Google Scholar] [CrossRef] [PubMed]
    47. Kampmann, C.; Perrin, A.; Beck, M. Effectiveness of agalsidase alfa enzyme replacement in Fabry disease: Cardiac outcomes after 10 years’ treatment. Orphanet J. Rare Dis. 2015, 10, 125. [Google Scholar] [CrossRef] [PubMed]
    48. Hughes, D.A.; Romero, M.A.B.; Hollak, C.E.M.; Giugliani, R.; Deegan, P.B. Response of women with Fabry disease to enzyme replacement therapy: Comparison with men, using data from FOS—the Fabry Outcome Survey. Mol. Genet. Metab. 2011, 103, 207–214. [Google Scholar] [CrossRef] [PubMed]
    49. Pastores, G.M.; Boyd, E.; Crandall, K.; Whelan, A.; Piersall, L.; Barnett, N. Safety and pharmacokinetics of agalsidase alfa in patients with Fabry disease and end-stage renal disease. Nephrol. Dial. Transplant. 2007, 22, 1920–1925. [Google Scholar] [CrossRef] [PubMed]
    50. Ramaswami, U.; Parini, R.; Kampmann, C.; Beck, M. Safety of agalsidase alfa in patients with Fabry disease under 7 years. Acta Paediatr. 2010, 100, 605–611. [Google Scholar] [CrossRef] [PubMed]
    51. Ries, M.; Clarke, J.T.; Whybra, C.; Timmons, M.; Robinson, C.; Schlaggar, B.L.; Pastores, G.; Lien, Y.H.; Kampmann, C.; Brady, R.O.; et al. Enzyme-replacement therapy with agalsidase alfa in children with Fabry disease. Pediatrics 2006, 118, 924–932. [Google Scholar] [CrossRef] [PubMed]
    52. Sasa, H.; Nagao, M.; Kino, K. Safety and effectiveness of enzyme replacement therapy with agalsidase alfa in patients with Fabry disease: Post-marketing surveillance in Japan. Mol. Genet. Metab. 2019, 126, 448–459. [Google Scholar] [CrossRef] [PubMed]
    53. Schiffmann, R.; Askari, H.; Timmons, M.; Robinson, C.; Benko, W.; Brady, R.O.; Ries, M. Weekly enzyme replacement therapy may slow decline of renal function in patients with Fabry disease who are on long-term biweekly dosing. J. Am. Soc. Nephrol. 2007, 18, 1576–1583. [Google Scholar] [CrossRef] [PubMed]
    54. Schiffmann, R.; Ries, M.; Timmons, M.; Flaherty, J.T.; Brady, R.O. Long-term therapy with agalsidase alfa for Fabry disease: Safety and effects on renal function in a home infusion setting. Nephrol. Dial. Transplant. 2006, 21, 345–354. [Google Scholar] [CrossRef] [PubMed]
    55. Schiffmann, R.; Pastores, G.M.; Lien, Y.-H.H.; Castaneda, V.; Chang, P.; Martin, R.; Wijatyk, A. Agalsidase alfa in pediatric patients with Fabry disease: A 6.5-year open-label follow-up study. Orphanet J. Rare Dis. 2014, 9, 169. [Google Scholar] [CrossRef] [PubMed]
    56. Schiffmann, R.; Ries, M.; Blankenship, D.; Nicholls, K.; Mehta, A.; Clarke, J.T.R.; Steiner, R.D.; Beck, M.; Barshop, B.A.; Rhead, W.; et al. Changes in plasma and urine globotriaosylceramide levels do not predict Fabry disease progression over 1 year of agalsidase alfa. Genet. Med. 2013, 15, 983–989. [Google Scholar]
    57. Schiffmann, R.; Swift, C.; Wang, X.; Blankenship, D.; Ries, M. A prospective 10-year study of individualized, intensified enzyme replacement therapy in advanced Fabry disease. J. Inherit. Metab. Dis. 2015, 38, 1129–1136. [Google Scholar] [CrossRef] [PubMed]
    58. Schwarting, A.; Dehout, F.; Beck, M.; Mehta, A.; Sunder-Plassmann, G.; Bodamer, O.; European FOS Investigators. Enzyme replacement therapy and renal function in 201 patients with Fabry disease. Clin. Nephrol. 2006, 66, 77–84. [Google Scholar] [PubMed]
    59. Schwarting, A.; Sunder-Plassmann, G.; Mehta, A.; Beck, M. Effect of enzyme replacement therapy with agalsidase alfa on renal function in patients with Fabry disease: Data from FOS—The Fabry Outcome Survey. In Fabry Disease: Perspectives from 5 Years of FOS; Adam, M.P., Feldman, J., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Amemiya, A., Eds.; Oxford PharmaGenesis: Oxford, UK, 2006; p. 38. [Google Scholar]
    60. Thofehrn, S.; Netto, C.; Cecchin, C.; Burin, M.; Matte, U.; Brustolin, S.; Nunes, A.C.F.; Coelho, J.; Tsao, M.; Jardim, L.; et al. Kidney function and 24-hour proteinuria in patients with Fabry disease during 36 months of agalsidase alfa enzyme replacement therapy: A Brazilian experience. Ren. Fail. 2009, 31, 773–778. [Google Scholar] [CrossRef] [PubMed]
    61. Tsuboi, K.; Yamamoto, H. Efficacy and safety of enzyme-replacement-therapy with agalsidase alfa in 36 treatment-naive Fabry disease patients. BMC Pharmacol. Toxicol. 2017, 18, 43. [Google Scholar] [CrossRef] [PubMed]
    62. West, M.; Nicholls, K.; Mehta, A.; Clarke, J.T.; Steiner, R.; Beck, M.; Barshop, B.A.; Rhead, W.; Mensah, R.; Ries, M.; et al. Agalsidase alfa and kidney dysfunction in Fabry disease. J. Am. Soc. Nephrol. 2009, 20, 1132–1139. [Google Scholar] [CrossRef] [PubMed]
    63. Whybra, C.; Miebach, E.; Mengel, E.; Gal, A.; Baron, K.; Beck, M.; Kampmann, C. A 4-year study of the efficacy and tolerability of enzyme replacement therapy with agalsidase alfa in 36 women with Fabry disease. Genet. Med. 2009, 11, 441–449. [Google Scholar] [CrossRef] [PubMed]
    64. Kleinert, J.; Dehout, F.; Schwarting, A.; de Lorenzo, A.; Ricci, R.; Kampmann, C.; Beck, M.; Ramaswami, U.; Linhart, A.; Gal, A.; et al. Prevalence of uncontrolled hypertension in patients with Fabry disease. Am. J. Hypertens. 2006, 19, 782–787. [Google Scholar] [CrossRef] [PubMed]
    65. Mehta, A.; Beck, M.; Elliott, P.; Giugliani, R.; Linhart, A.; Sunder-Plassmann, G.; Schiffmann, R.; Barbey, F.; Ries, M.; Clarke, J. Enzyme replacement therapy with agalsidase alfa in patients with Fabry’s disease: An analysis of registry data. Lancet 2009, 374, 1986–1996. [Google Scholar] [CrossRef] [PubMed]
    66. Ramaswami, U.; Parini, R.; Pintos-Morell, G.; Kalkum, G.; Kampmann, C.; Beck, M. Fabry disease in children and response to enzyme replacement therapy: Results from the Fabry Outcome Survey. Clin. Genet. 2011, 81, 485–490. [Google Scholar] [CrossRef] [PubMed]
    67. Benichou, B.; Goyal, S.; Sung, C.; Norfleet, A.M.; O’Brien, F. A retrospective analysis of the potential impact of IgG antibodies to agalsidase beta on efficacy during enzyme replacement therapy for Fabry disease. Mol. Genet. Metab. 2009, 96, 4–12. [Google Scholar] [CrossRef] [PubMed]
    68. Breunig, F.; Weidemann, F.; Strotmann, J.; Knoll, A.; Wanner, C. Clinical benefit of enzyme replacement therapy in Fabry disease. Kidney Int. 2006, 69, 1216–1221. [Google Scholar] [CrossRef] [PubMed]
    69. Veloso, V.S.P.; Veloso, M.; Pereira, E.; Barreto, F.; Hagemann, R. Algalsidase-beta stabilizes renal and cardiac involvement: A follow-up study of a Brazilian Fabry disease family. Nephrol. Dial. Transplant. 2023, 38 (Suppl. S1), 5209. [Google Scholar] [CrossRef]
    70. Burlina, A.P.; Mauer, M.; Cabrera, G.H.; Jefferies, J.L.; Yang, M.; Ponce, E.; Brand, E.; Feldt-Rasmussen, U.; Germain, P. Clinical outcomes in young patients with Fabry disease who initiated agalsidase beta treatment before 30 years of age. J. Inherit. Metab. Dis. 2019, 42 (Suppl. S1), 233–234. [Google Scholar]
    71. Dutra-Clarke, M.; Tapia, D.; Curtin, E.; Ruenger, D.; Lakatos, A.; Alandy-Dy, Z.; Freedkin, L.; Hall, K.; Ercelen, N.; Alandy-Dy, J.; et al. Variable clinical features of patients with Fabry disease and outcome of enzyme replacement therapy. Mol. Genet. Metab. 2021, 132 (Suppl. S2), S36. [Google Scholar] [CrossRef]
    72. Goicoechea, M.; Gomez-Preciado, F.; Benito, S.; Torras, J.; Torra, R.; Huerta, A.; Restrepo, A.; Ugalde, J.; Astudillo, D.; Agraz, I.; et al. Predictors of long-term outcome in a Spanish cohort of patients with Fabry disease on enzyme replacement therapy. Nephrol. Dial. Transplant. 2020, 35 (Suppl. S3), iii363. [Google Scholar] [CrossRef]
    73. Hwang, S.; Lee, B.H.; Kim, W.-S.; Kim, D.-S.; Cheon, C.K.; Lee, C.H.; Choi, Y.; Choi, J.-H.; Kim, J.H.; Yoo, H.-W. A phase II, multicenter, open-label trial to evaluate the safety and efficacy of ISU303 (Agalsidase beta) in patients with Fabry disease. Medicine 2022, 101, e30345. [Google Scholar] [CrossRef] [PubMed]
    74. Lubanda, J.C.; Anijalg, E.; Bzduch, V.; Thurberg, B.L.; Benichou, B.; Tylki-Szymanska, A. Evaluation of a low dose, after a standard therapeutic dose, of agalsidase beta during enzyme replacement therapy in patients with Fabry disease. Genet. Med. 2009, 11, 256–264. [Google Scholar] [CrossRef] [PubMed]
    75. Ortiz, A.; Mauer, M.; Ponce, E.; Yang, M.; Gudigava, B.; Hong, G.R. Stabilization of kidney function decline and cardiomyopathy in male patients with classic Fabry disease: A pre-vs. post-agalsidase beta treatment Fabry Registry analysis. Nephrol. Dial. Transplant. 2021, 36 (Suppl. S1), i110–i111. [Google Scholar] [CrossRef]
    76. Ramaswami, U.; Bichet, D.G.; Clarke, L.A.; Dostalova, G.; Fainboim, A.; Fellgiebel, A.; Forcelini, C.M.; Haack, K.A.; Hopkin, R.J.; Mauer, M.; et al. Low-dose agalsidase beta treatment in male pediatric patients with Fabry disease: A 5-year randomized controlled trial. Mol. Genet. Metab. 2019, 127, 86–94. [Google Scholar] [CrossRef] [PubMed]
    77. Wanner, C.; Feldt-Rasmussen, U.; Jovanovic, A.; Linhart, A.; Yang, M.; Ponce, E.; Brand, E.; Germain, D.P.; Hughes, D.A.; Jefferies, J.L.; et al. Cardiomyopathy and kidney function in agalsidase beta-treated female Fabry patients: A pre-treatment vs. post-treatment analysis. ESC Heart Fail. 2020, 7, 825–834. [Google Scholar] [CrossRef] [PubMed]
    78. Warnock, D.G.; Ortiz, A.; Mauer, M.; Linthorst, G.E.; Oliveira, J.P.; Serra, A.L.; Maródi, L.; Mignani, R.; Vujkovac, B.; Beitner-Johnson, D.; et al. Renal outcomes of agalsidase beta treatment for Fabry disease: Role of proteinuria and timing of treatment initiation. Nephrol. Dial. Transplant. 2012, 27, 1042–1049. [Google Scholar] [CrossRef] [PubMed]
    79. Warnock, D.G.; Thomas, C.P.; Vujkovac, B.; Campbell, R.C.; Charrow, J.; A Laney, D.; Jackson, L.L.; Wilcox, W.R.; Wanner, C. Antiproteinuric therapy and Fabry nephropathy: Factors associated with preserved kidney function during agalsidase-beta therapy. J. Med. Genet. 2015, 52, 860–866. [Google Scholar] [CrossRef] [PubMed]
    80. Weidemann, F.; Niemann, M.; Störk, S.; Breunig, F.; Beer, M.; Sommer, C.; Herrmann, S.; Ertl, G.; Wanner, C. Long-term outcome of enzyme-replacement therapy in advanced Fabry disease: Evidence for disease progression towards serious complications. J. Intern. Med. 2013, 274, 331–341. [Google Scholar] [CrossRef] [PubMed]
    81. Wraith, J.E.; Tylki-Szymanska, A.; Guffon, N.; Lien, Y.H.; Tsimaratos, M.; Vellodi, A.; Germain, D.P. Safety and efficacy of enzyme replacement therapy with agalsidase beta: An international, open-label study in pediatric patients with Fabry disease. J. Pediatr. 2008, 152, 563–570.e1. [Google Scholar] [CrossRef] [PubMed]
    82. Germain, D.P.; Charrow, J.; Desnick, R.J.; Guffon, N.; Kempf, J.; Lachmann, R.H.; Lemay, R.; E Linthorst, G.; Packman, S.; Scott, C.R.; et al. Ten-year outcome of enzyme replacement therapy with agalsidase beta in patients with Fabry disease. J. Med. Genet. 2015, 52, 353–358. [Google Scholar] [CrossRef] [PubMed]
    83. Chen, N.; Ren, H.; Zhang, W.; Ouyang, Y.; Xu, H.; Li, B.; Feng, Q. A phase 4, open-label, multicenter prospective study of the safety and efficacy of agalsidase beta in Chinese patients with Fabry disease. Nephrol. Dial. Transplant. 2024, 39 (Suppl. S1), 636. [Google Scholar] [CrossRef]
    84. Goicoechea, M.; Gomez-Preciado, F.; Benito, S.; Torras, J.; Torra, R.; Huerta, A.; Restrepo, A.; Ugalde, J.; Astudillo, D.E.; Agraz, I.; et al. Predictors of outcome in a Spanish cohort of patients with Fabry disease on enzyme replacement therapy. Nefrologia 2021, 41, 652–660. [Google Scholar] [CrossRef] [PubMed]
    85. Lin, H.-Y.; Huang, Y.-H.; Liao, H.-C.; Liu, H.-C.; Hsu, T.-R.; Shen, C.-I.; Li, S.-T.; Li, C.-F.; Lee, L.-H.; Lee, P.-C.; et al. Clinical observations on enzyme replacement therapy in patients with Fabry disease and the switch from agalsidase beta to agalsidase alfa. J. Chin. Med Assoc. 2014, 77, 190–197. [Google Scholar] [CrossRef] [PubMed]
    86. Lin, H.-Y.; Liu, H.-C.; Huang, Y.-H.; Liao, H.-C.; Hsu, T.-R.; Shen, C.-I.; Li, S.-T.; Li, C.-F.; Lee, L.-H.; Lee, P.-C.; et al. Effects of enzyme replacement therapy i for cardiac-type Fabry patients with a Chinese hotspot late-onset Fabry mutation (IVS4+919G>A). BMJ Open 2013, 3, e003146. [Google Scholar] [CrossRef] [PubMed]
    87. Madsen, C.V.; Granqvist, H.; Petersen, J.H.; Rasmussen, Å.K.; Lund, A.M.; Oturai, P.; Sørensen, S.S.; Feldt-Rasmussen, U. Age-related renal function decline in Fabry disease patients on enzyme replacement therapy: A longitudinal cohort study. Nephrol. Dial. Transplant. 2019, 34, 1525–1533. [Google Scholar] [CrossRef] [PubMed]
    88. Miwa, K.; Sakai, N.; Yagita, Y.; Takeuchi, M.; Todo, K.; Sakaguchi, M.; Mochizuki, H. Basilar artery diameter is associated with cerebral small vessel disease and the duration of enzyme-replacement therapy in Fabry disease. Stroke. 2018, 49 (Suppl. 1), WP388. [Google Scholar] [CrossRef]
    89. Miwa, K.; Yagita, Y.; Sakaguchi, M.; Kitagawa, K.; Sakai, N.; Mochizuki, H. Effect of enzyme replacement therapy on basilar artery diameter in male patients with Fabry disease. Stroke 2019, 50, 1010–1012. [Google Scholar] [CrossRef] [PubMed]
    90. Rombach, S.M.; Smid, B.E.; Bouwman, M.G.; Linthorst, G.E.; Dijkgraaf, M.G.; Hollak, C.E. Long term enzyme replacement therapy for Fabry disease: Effectiveness on kidney, heart and brain. Orphanet J. Rare Dis. 2013, 8, 47. [Google Scholar] [CrossRef] [PubMed]
    91. Sirrs, S.; Bichet, D.; Casey, R.; Clarke, J.; Lemoine, K.; Doucette, S.; West, M. Outcomes of patients treated through the Canadian Fabry disease initiative. Mol. Genet. Metab. 2014, 111, 499–506. [Google Scholar] [CrossRef] [PubMed]
    92. Skrunes, R.; Tøndel, C.; Leh, S.; Larsen, K.K.; Houge, G.; Davidsen, E.S.; Hollak, C.; van Kuilenburg, A.B.; Vaz, F.M.; Svarstad, E. Long-term dose-dependent agalsidase effects on kidney histology in Fabry disease. Clin. J. Am. Soc. Nephrol. 2017, 12, 1470–1479. [Google Scholar] [CrossRef] [PubMed]
    93. Talbot, A.S.; Lewis, N.T.; Nicholls, K.M. Cardiovascular outcomes in Fabry disease are linked to severity of chronic kidney disease. Heart 2015, 101, 287–293. [Google Scholar] [CrossRef] [PubMed]
    94. Bernat, J.; Holida, M.D.; Longo, N.; Goker-Alpan, O.; Wallace, E.; Deegan, P.B.; Tøndel, C.; Eyskens, F.J.; Feldt-Rasmussen, U.; Hughes, D.; et al. Long-term safety and efficacy of pegunigalsidase alfa administered every 4 weeks in patients with Fabry disease: Two-year interim results from the ongoing phase 3 BRIGHT51 open-label extension study. Mol. Genet. Metab. 2023, 138, 107027. [Google Scholar] [CrossRef]
    95. Hughes, D.; Gonzalez, D.; Maegawa, G.; Bernat, J.A.; Holida, M.; Giraldo, P.; Atta, M.G.; Chertkoff, R.; Alon, S.; Almon, E.B.; et al. Long-term safety and efficacy of pegunigalsidase alfa: A multicenter 6-year study in adult patients with Fabry disease. Genet. Med. 2023, 25, 100968. [Google Scholar] [CrossRef] [PubMed]
    96. Bernat, J.; Holida, M.; Longo, N.; Goker-Alpan, O.; Wallace, E.; Deegan, P.; Nedd, K.; Tondel, C.; Eyskens, F.; Feldt-Rasmussen, U.; et al. eP149: Safety and efficacy of pegunigalsidase alfa, every 4 weeks, in Fabry disease: Results from the phase 3, open-label, BRIGHT study. Genet. Med. 2022, 24 (Suppl. S3), S91–S92. [Google Scholar] [CrossRef]
    97. West, M.L.; Hughes, D.; Sunder-Plassmann, G.; Jovanovic, A.; Brand, E.; Bichet, D.G.; Pisani, A.; Nowak, A.; Torra, R.; Khan, A.; et al. FollowME Fabry Pathfinders Registry: Renal effectiveness in a cohort of patients on migalastat treatment for at least three years. J. Am. Soc. Nephrol. 2023, 34 (Suppl. S11), 952. [Google Scholar] [CrossRef]
    98. Germain, D.P.; Hughes, D.A.; Nicholls, K.; Bichet, D.G.; Giugliani, R.; Wilcox, W.R.; Feliciani, C.; Shankar, S.P.; Ezgu, F.; Amartino, H.; et al. Treatment of Fabry’s disease with the pharmacologic chaperone migalastat. N. Engl. J. Med. 2016, 375, 545–555. [Google Scholar] [CrossRef] [PubMed]
    99. Germain, D.P.; Nicholls, K.; Giugliani, R.; Bichet, D.G.; Hughes, D.A.; Barisoni, L.M.; Colvin, R.B.; Jennette, J.C.; Skuban, N.; Castelli, J.P.; et al. Efficacy of the pharmacologic chaperone migalastat in a subset of male patients with the classic phenotype of Fabry disease and migalastat-amenable variants: Data from the phase 3 randomized, multicenter, double-blind clinical trial and extension study. Genet. Med. 2019, 21, 1987–1997. [Google Scholar] [CrossRef] [PubMed]
    100. Giugliani, R.; Waldek, S.; Germain, D.; Nicholls, K.; Bichet, D.; Simosky, J.; Bragat, A.; Castelli, J.; Benjamin, E.; Boudes, P. A Phase 2 study of migalastat hydrochloride in females with Fabry disease: Selection of population, safety and pharmacodynamic effects. Mol. Genet. Metab. 2013, 109, 86–92. [Google Scholar] [CrossRef] [PubMed]
    101. Hopkin, R.J.; Bichet, D.G.; Sunder-Plassmann, G.; Nicholls, K.; Olivotto, I.; Giugliani, R.; Krusinska, E.; Veleva-Rotse, B.; Hughes, D. Long-term multisystemic efficacy with migalastat in ERT-naive and ERT-experienced patients with amenable GLA variants. Mol. Genet. Metab. 2022, 135 (Suppl. S2), S56. [Google Scholar]
    102. Khan, A.; Barber, D.L.; Huang, J.; Rupar, C.A.; Rip, J.W.; Auray-Blais, C.; Boutin, M.; O’Hoski, P.; Gargulak, K.; McKillop, W.M.; et al. Lentivirus-mediated gene therapy for Fabry disease. Nat. Commun. 2021, 12, 1178. [Google Scholar] [CrossRef] [PubMed]
    103. Lenders, M.; Nordbeck, P.; Kurschat, C.; Eveslage, M.; Karabul, N.; Kaufeld, J.; Hennermann, J.B.; Patten, M.; Cybulla, M.; Müntze, J.; et al. Treatment of Fabry disease management with migalastat—outcome from a prospective 24 months observational multicenter study (FAMOUS). Eur. Heart J. Cardiovasc. Pharmacother. 2022, 8, 272–281. [Google Scholar] [CrossRef] [PubMed]
    104. Lenders, M.; Nordbeck, P.; Kurschat, C.; Karabul, N.; Kaufeld, J.; Hennermann, J.B.; Patten, M.; Cybulla, M.; Müntze, J.; Üçeyler, N.; et al. Treatment of Fabry’s disease with migalastat: Outcome from a prospective observational multicenter study (FAMOUS). Clin. Pharmacol. Ther. 2020, 108, 326–337. [Google Scholar] [CrossRef] [PubMed]
    105. Orsborne, C.; Thompson, L.; Jovanovic, A. A retrospective outcome analysis of chaperone therapy in Fabry disease: Clinical outcomes after the first year of therapy—A single centre experience. Mol. Genet. Metab. 2020, 129 (Suppl. S2), S123. [Google Scholar] [CrossRef]
    106. Ramaswami, U.; Wilcox, W.; Hopkin, R.J.; Yang, H.; Jiang, H.; Lengoc, V. Migalastat HCl 150mg every other day is well-tolerated and efficacious in adolescent patients with Fabry disease. Mol. Genet. Metab. 2022, 135 (Suppl. S2), S104. [Google Scholar] [CrossRef]
    107. Schiffmann, R.; Goker-Alpan, O.; Holida, M.; Giraldo, P.; Barisoni, L.; Colvin, R.B.; Jennette, C.J.; Maegawa, G.; Boyadjiev, S.A.; Gonzalez, D.; et al. Pegunigalsidase alfa, a novel PEGylated enzyme replacement therapy for Fabry disease, provides sustained plasma concentrations and favorable pharmacodynamics: A 1-year Phase 1/2 clinical trial. J. Inherit. Metab. Dis. 2019, 42, 534–544. [Google Scholar] [CrossRef] [PubMed]
    108. Skuban, N.; Feldt-Rasmussen, U.; Giugliani, R.; Germain, D.P.; Hughes, D.A.; Wilcox, W.R.; Schiffmann, R.; Bichet, D.G.; Jovanovic, A.; Bratkovic, D.; et al. Efficacy and safety of migalastat, an oral pharmacologic chaperone for Fabry disease: Results from two randomized phase 3 studies, FACETS and ATTRACT. Nephron 2017, 136, 175. [Google Scholar]
    109. Hughes, D.; Sunder-Plassmann, G.; Jovanovic, A.; Brand, E.; West, M.L.; Bichet, D.G.; Pisani, A.; Nowak, A.; Torra, R.; Khan, A.; et al. FollowME Fabry Pathfinders registry: Renal effectiveness in a multi-national, multi-center cohort of patients on migalastat treatment for at least three years. Mol. Genet. Metab. 2023, 138, 107159. [Google Scholar] [CrossRef]
    110. Bichet, D.G.; Torra, R.; Wallace, E.; Hughes, D.; Giugliani, R.; Skuban, N.; Krusinska, E.; Feldt-Rasmussen, U.; Schiffmann, R.; Nicholls, K. Long-term follow-up of renal function in patients treated with migalastat for Fabry disease. Mol. Genet. Metab. Rep. 2021, 28, 100786. [Google Scholar] [CrossRef] [PubMed]
    111. Torra, R.; Germain, D.; Bichet, D.; Schiffmann, R.; Yu, J.; Castelli, J.; Skuban, N.; Barth, J. Clinical outcomes with migalastat in patients with Fabry disease based on degree of renal impairment: Results from phase 3 trials. Nephrol. Dial. Transplant. 2018, 33 (Suppl. S1), i346–i356. [Google Scholar] [CrossRef]
    112. Beck, M.; Hughes, D.; Kampmann, C.; Larroque, S.; Mehta, A.; Pintos-Morell, G.; Ramaswami, U.; West, M.; Wijatyk, A.; Giugliani, R. Long-term effectiveness of agalsidase alfa enzyme replacement in Fabry disease: A Fabry Outcome Survey analysis. Mol. Genet. Metab. Rep. 2015, 3, 21–27. [Google Scholar] [CrossRef] [PubMed]
    113. Cybulla, M.; Walter, K.N.; Schwarting, A.; Divito, R.; Feriozzi, S.; Sunder-Plassmann, G. Kidney transplantation in patients with Fabry disease. Transpl. Int. 2009, 22, 475–481. [Google Scholar] [CrossRef] [PubMed]
    114. Cybulla, M.; West, M.; Nicholls, K.; Torras, J.; Neumann, P.; Sunder-Plassmann, G.; Feriozzi, S.; on behalf of the Fabry Outcome Survey Renal Working Group. Efficacy and safety of enzyme replacement therapy (ERT) in a cohort of kidney transplant recipients with Fabry disease. Nephrol. Dial. Transplant. 2014, 29 (Suppl. S3), iii349. [Google Scholar]
    115. Cybulla, M.; West, M.; Nicholls, K.; Torras, J.; Sunder-Plassmann, G.; Feriozzi, S. Effectiveness and safety of enzyme replacement therapy (ERT) in a cohort of kidney transplant recipients with Fabry disease. Nephrol. Dial. Transplant. 2013, 28 (Suppl. S1), i287. [Google Scholar]
    116. Dehout, F.; Schwarting, A.; Beck, M.; Mehta, A.; Ricci, R.; Widmer, U. Effects of enzyme replacement therapy with agalsidase alfa on glomerular filtration rate in patients with Fabry disease: Preliminary data. Acta Paediatr. 2003, 92 (Suppl. S443), 14–15. [Google Scholar] [CrossRef] [PubMed]
    117. Eng, C.M.; Banikazemi, M.; Gordon, R.E.; Goldman, M.; Phelps, R.; Kim, L.; Gass, A.; Winston, J.; Dikman, S.; Fallon, J.T.; et al. A phase 1/2 clinical trial of enzyme replacement in Fabry disease: Pharmacokinetic, substrate clearance, and safety studies. Am. J. Hum. Genet. 2001, 68, 711–722. [Google Scholar] [CrossRef] [PubMed]
    118. Goláň, L.; Goker-Alpan, O.; Holida, M.; Kantola, I.; Klopotowski, M.; Kuusisto, J.; Linhart, A.; Musial, J.; Nicholls, K.; Rodríguez, D.G.; et al. Evaluation of the efficacy and safety of three dosing regimens of agalsidase alfa enzyme replacement therapy in adults with Fabry disease. Drug Des. Dev. Ther. 2015, 9, 3435–3444. [Google Scholar] [CrossRef] [PubMed]
    119. Guérard, N.; Oder, D.; Nordbeck, P.; Zwingelstein, C.; Morand, O.; Welford, R.W.; Dingemanse, J.; Wanner, C. Lucerastat, an iminosugar for substrate reduction therapy: Tolerability, pharmacodynamics, and pharmacokinetics in patients with Fabry disease on enzyme replacement. Clin. Pharmacol. Ther. 2018, 103, 703–711. [Google Scholar] [CrossRef] [PubMed]
    120. Hughes, D.; Romero, M.B.; Gurevich, A.; Engrand, P.; Giugliani, R. Menarche, menopause, and pregnancy data in untreated/agalsidase alfa-treated females in the Fabry Outcome Survey. Twin Res. Hum. Genet. 2019, 21, 439. [Google Scholar] [CrossRef]
    121. Hughes, D.A.; Elliott, P.M.; Shah, J.; Zuckerman, J.; Coghlan, G.; Brookes, J.; Mehta, A.B. Effects of enzyme replacement therapy on the cardiomyopathy of Anderson-Fabry disease: A randomised, double-blind, placebo-controlled clinical trial of agalsidase alfa. Heart 2008, 94, 153–158. [Google Scholar] [CrossRef] [PubMed]
    122. Jovanovich, A.; Schiffmann, R.; Nicholls, K.; Bichet, D.; Feldt-Rasmussen, U.; Jovanovic, A.; Hughes, D.; Jain, V.; Castelli, J.; Skuban, N.; et al. Efficacy and safety of migalastat, an oral pharmacological chaperone for Fabry disease: Renal findings from two randomized phase 3 studies (facets and attract). J. Inborn Errors Metab. Screen. 2017, 5, 356. [Google Scholar]
    123. Prabakaran, T.; Birn, H.; Bibby, B.M.; Regeniter, A.; Sørensen, S.S.; Feldt-Rasmussen, U.; Nielsen, R.; Christensen, E.I. Long-term enzyme replacement therapy is associated with reduced proteinuria and preserved proximal tubular function in women with Fabry disease. Nephrol. Dial. Transplant. 2014, 29, 619–625. [Google Scholar] [CrossRef] [PubMed]
    124. Schiffmann, R.; Bichet, D.; Germain, D.; Giugliani, R.; Hughes, D.; Nicholls, K.; Wilcox, W.; Williams, H.; Yu, J.; Castelli, J.; et al. Effects of long-term migalastat treatment on renal function by baseline proteinuria in patients (PTS) with Fabry disease. Nephrol. Dial. Transplant. 2018, 33 (Suppl. S1), i346–i347. [Google Scholar] [CrossRef]
    125. Wallace, E.; Goker-Alpan, O.; Alon, S.; Chertkoff, R.; Almon, E.; Rocco, R.; Warnock, D.G.; Longo, N. Safety and efficacy of pegunigalsidase alfa vs agalsidase beta on renal function in Fabry disease: 24-month results from the phase III randomized, double-blind, BALANCE study. Nephron 2022, 146 (Suppl. S1), 39. [Google Scholar]
    126. Wilcox, W.R.; Banikazemi, M.; Guffon, N.; Waldek, S.; Lee, P.; Linthorst, G.E.; Desnick, R.J.; Germain, D.P.; International Fabry Disease Study Group. Long-term safety and efficacy of enzyme replacement therapy for Fabry disease. Am. J. Hum. Genet. 2004, 75, 65–74. [Google Scholar] [CrossRef] [PubMed]
    127. Wallace, E.L.; Goker-Alpan, O.; Wilcox, W.R.; Holida, M.; Bernat, J.; Longo, N.; Linhart, A.; A Hughes, D.; Hopkin, R.J.; Tøndel, C.; et al. 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. J. Med. Genet. 2023, 61, 520–530. [Google Scholar] [CrossRef] [PubMed]
    128. Goker-Alpan, O.; Nedd, K.; Shankar, S.P.; Lien, Y.-H.H.; Weinreb, N.; Wijatyk, A.; Chang, P.; Martin, R. Effect and tolerability of agalsidase alfa in patients with Fabry disease who were treatment naive or formerly treated with agalsidase beta or agalsidase alfa. JIMD Rep. 2015, 23, 7–15. [Google Scholar] [PubMed]
    129. Hughes, D.A.; Nicholls, K.; Shankar, S.P.; Sunder-Plassmann, G.; Koeller, D.; Nedd, K.; Vockley, G.; Hamazaki, T.; Lachmann, R.; Ohashi, T.; et al. Oral pharmacological chaperone migalastat compared with enzyme replacement therapy in Fabry disease: 18-month results from the randomised phase III ATTRACT study. J. Med. Genet. 2017, 54, 288–296. [Google Scholar] [CrossRef] [PubMed]
    130. Narita, I.; Ohashi, T.; Sakai, N.; Hamazaki, T.; Skuban, N.; Castelli, J.P.; Lagast, H.; Barth, J.A. Efficacy and safety of migalastat in a Japanese population: A subgroup analysis of the ATTRACT study. Clin. Exp. Nephrol. 2020, 24, 157–166. [Google Scholar] [CrossRef] [PubMed]
    131. Krämer, J.; Lenders, M.; Canaan-Kühl, S.; Nordbeck, P.; Üçeyler, N.; Blaschke, D.; Duning, T.; Reiermann, S.; Stypmann, J.; Brand, S.-M.; et al. Fabry disease under enzyme replacement therapy—new insights in efficacy of different dosages. Nephrol. Dial. Transplant. 2018, 33, 1362–1372. [Google Scholar] [CrossRef] [PubMed]
    132. Lenders, M.; Canaan-Kühl, S.; Krämer, J.; Duning, T.; Reiermann, S.; Sommer, C.; Stypmann, J.; Blaschke, D.; Üçeyler, N.; Hense, H.-W.; et al. Patients with Fabry disease after enzyme replacement therapy dose reduction and switch-2-year follow-up. J. Am. Soc. Nephrol. 2016, 27, 952–962. [Google Scholar] [CrossRef] [PubMed]
    133. Lenders, M.; Nordbeck, P.; Canaan-Kühl, S.; Kreul, L.; Duning, T.; Lorenz, L.; Pogoda, C.; Brand, S.-M.; Wanner, C.; Brand, E. Treatment switch in Fabry disease—a matter of dose? J. Med. Genet. 2021, 58, 342–350. [Google Scholar] [CrossRef] [PubMed]
    134. Muentze, J.; Gensler, D.; Salinger, T.; Oder, D.; Wanner, C.; Frantz, S.; Nordbeck, P. Treatment of cardiac manifestations in Fabry disease with the oral drug migalastat: First 12 months results from a cohort of amenable all-comers. Eur. Heart J. 2018, 39 (Suppl. S1), 460. [Google Scholar] [CrossRef]
    135. Müntze, J.; Gensler, D.; Maniuc, O.; Liu, D.; Cairns, T.; Oder, D.; Hu, K.; Lorenz, K.; Frantz, S.; Wanner, C.; et al. Oral chaperone therapy migalastat for treating Fabry disease: Enzymatic response and serum biomarker changes after 1 year. Clin. Pharmacol. Ther. 2019, 105, 1224–1233. [Google Scholar] [CrossRef] [PubMed]
    136. Tsuboi, K.; Yamamoto, H. Clinical observation of patients with Fabry disease after switching from agalsidase beta (Fabrazyme) to agalsidase alfa (Replagal). Genet. Med. 2012, 14, 779–786. [Google Scholar] [CrossRef] [PubMed]
    137. Tsuboi, K.; Yamamoto, H. Clinical course of patients with Fabry disease who were switched from agalsidase-beta to agalsidase-alpha. Genet. Med. 2014, 16, 766–772. [Google Scholar] [CrossRef] [PubMed]
    138. Weidemann, F.; Krämer, J.; Duning, T.; Lenders, M.; Canaan-Kühl, S.; Krebs, A.; González, H.G.; Sommer, C.; Üçeyler, N.; Niemann, M.; et al. Patients with Fabry disease after enzyme replacement therapy dose reduction versus treatment switch. J. Am. Soc. Nephrol. 2014, 25, 837–849. [Google Scholar] [CrossRef] [PubMed]
    139. Linhart, A.; Dostálová, G.; Nicholls, K.; West, M.L.; Tøndel, C.; Jovanovic, A.; Giraldo, P.; Vujkovac, B.; Geberhiwot, T.; Brill-Almon, E.; et al. Safety and efficacy of pegunigalsidase alfa in patients with Fabry disease who were previously treated with agalsidase alfa: Results from BRIDGE, a phase 3 open-label study. Orphanet J. Rare Dis. 2023, 18, 332. [Google Scholar] [CrossRef] [PubMed]
    140. Riccio, E.; Zanfardino, M.; Ferreri, L.; Santoro, C.; Cocozza, S.; Capuano, I.; Imbriaco, M.; Feriozzi, S.; Pisani, A.; AFFIINITY Group. Switch from enzyme replacement therapy to oral chaperone migalastat for treating Fabry disease: Real-life data. Eur. J. Hum. Genet. 2020, 28, 1662–1668. [Google Scholar] [CrossRef] [PubMed]
    141. Baehner, F.; Kampmann, C.; Whybra, C.; Miebach, E.; Wiethoff, C.M.; Beck, M. Enzyme replacement therapy in heterozygous females with Fabry disease: Results of a phase IIIB study. J. Inherit. Metab. Dis. 2003, 26, 617–627. [Google Scholar] [CrossRef] [PubMed]
    142. Hughes, D.A.; Deegan, P.B.; Romero, M.B.; Giugliani, R. Responses of females with Fabry disease to ERT: Comparison with males, using data from FOS—The Fabry outcome survey. Mol. Genet. Metab. 2009, 98, 81. [Google Scholar]
    143. Kampmann, C.; Linhart, A.; Devereux, R.B.; Schiffmann, R. Effect of agalsidase alfa replacement therapy on Fabry disease-related hypertrophic cardiomyopathy: A 12- to 36-month, retrospective, blinded echocardiographic pooled analysis. Clin. Ther. 2009, 31, 1966–1976. [Google Scholar] [CrossRef] [PubMed]
    144. Elliott, P.M.; Kindler, H.; Shah, J.S.; Sachdev, B.; Rimoldi, O.E.; Thaman, R.; Tome, M.T.; McKenna, W.J.; Lee, P.; Camici, P.G. Coronary microvascular dysfunction in male patients with Anderson-Fabry disease and the effect of treatment with alpha galactosidase A. Heart 2006, 92, 357–360. [Google Scholar] [CrossRef] [PubMed]
    145. Kalliokoski, R.J.; Kantola, I.; Kalliokoski, K.K.; Engblom, E.; Sundell, J.; Hannukainen, J.C.; Janatuinen, T.; Raitakari, O.T.; Knuuti, J.; Penttinen, M.; et al. The effect of 12-month enzyme replacement therapy on myocardial perfusion in patients with Fabry disease. J. Inherit. Metab. Dis. 2006, 29, 112–118. [Google Scholar] [CrossRef] [PubMed]
    146. Messalli, G.; Imbriaco, M.; Avitabile, G.; Russo, R.; Iodice, D.; Spinelli, L.; Dellegrottaglie, S.; Cademartiri, F.; Salvatore, M.; Pisani, A. Role of cardiac MRI in evaluating patients with Anderson-Fabry disease: Assessing cardiac effects of long-term enzyme replacement therapy. Radiol. Med. 2012, 117, 19–28. [Google Scholar] [CrossRef] [PubMed]
    147. Motwani, M.; Banypersad, S.; Woolfson, P.; Waldek, S. Enzyme replacement therapy improves cardiac features and severity of Fabry disease. Mol. Genet. Metab. 2012, 107, 197–202. [Google Scholar] [CrossRef] [PubMed]
    148. Pisani, A.; Spinelli, L.; Sabbatini, M.; Andreucci, M.V.; Procaccini, D.; Abbaterusso, C.; Pasquali, S.; Savoldi, S.; Comotti, C.; Cianciaruso, B. Enzyme replacement therapy in Fabry disease patients undergoing dialysis: Effects on quality of life and organ involvement. Am. J. Kidney Dis. 2005, 46, 120–127. [Google Scholar] [CrossRef] [PubMed]
    149. Wuest, W.; Machann, W.; Breunig, F.; Weidemann, F.; Koestler, H.; Hahn, D.; Wanner, C.; Beer, M. Right ventricular involvement in patients with Fabry disease and the effect of enzyme replacement therapy. Rofo 2011, 183, 1037–1042. [Google Scholar] [CrossRef] [PubMed]
    150. Pietilä-Effati, P.; Saarinen, J.T.; Loyttyniemi, E.; Saarenhovi, M.; Autio, R.; Kantola, I. Long-term effectiveness of enzyme replacement therapy in Fabry disease with the p.Arg227Ter (R227*) mutation. Mol. Genet. Metab. 2023, 138, 107270. [Google Scholar] [CrossRef]
    151. Huang, S.; Wang, J.; Zhang, W.; Gao, F.; Chen, Y.; Shui, W.; Xing, X.; Chen, S.; Mu, Y. Clinical study of left ventricular structure and function in patients with Anderson-Fabry disease before and after enzyme replacement therapy. J. Clin. Ultrasound 2024, 52, 20–29. [Google Scholar] [CrossRef] [PubMed]
    152. Hongo, K.; Ito, K.; Date, T.; Anan, I.; Inoue, Y.; Morimoto, S.; Ogawa, K.; Kawai, M.; Kobayashi, H.; Kobayashi, M.; et al. The beneficial effects of long-term enzyme replacement therapy on cardiac involvement in Japanese Fabry patients. Mol. Genet. Metab. 2018, 124, 143–151. [Google Scholar] [CrossRef] [PubMed]
    153. Kovacevic-Preradovic, T.; Zuber, M.; Jost, C.A.; Widmer, U.; Seifert, B.; Schulthess, G.; Fischer, A.; Jenni, R. Anderson-Fabry disease: Long-term echocardiographic follow-up under enzyme replacement therapy. Eur. J. Echocardiogr. 2008, 9, 729–735. [Google Scholar] [CrossRef] [PubMed]
    154. Lenders, M.; Stypmann, J.; Duning, T.; Schmitz, B.; Brand, S.M.; Brand, E. Serum-mediated inhibition of enzyme replacement therapy in Fabry disease. J. Am. Soc. Nephrol. 2016, 27, 256–264. [Google Scholar] [CrossRef] [PubMed]
    155. Liu, H.-C.; Lin, H.-Y.; Yang, C.-F.; Liao, H.-C.; Hsu, T.-R.; Lo, C.-W.; Chang, F.-P.; Huang, C.-K.; Lu, Y.-H.; Lin, S.-P.; et al. Globotriaosylsphingosine (lyso-Gb3) might not be a reliable marker for monitoring the long-term therapeutic outcomes of enzyme replacement therapy for late-onset Fabry patients with the Chinese hotspot mutation (IVS4+919G>A). Orphanet J. Rare Dis. 2014, 9, 111. [Google Scholar] [CrossRef] [PubMed]
    156. Nordin, S.; Kozor, R.; Vijapurapu, R.; Augusto, J.B.; Knott, K.D.; Captur, G.; Treibel, T.A.; Ramaswami, U.; Tchan, M.; Geberhiwot, T.; et al. Myocardial storage, inflammation, and cardiac phenotype in Fabry disease after one year of enzyme replacement therapy. Circ. Cardiovasc. Imaging 2019, 12, e009430. [Google Scholar] [CrossRef] [PubMed]
    157. Schmied, C.; Nowak, A.; Gruner, C.; Olinger, E.; Debaix, H.; Brauchlin, A.; Frank, M.; Reidt, S.; Monney, P.; Barbey, F.; et al. The value of ECG parameters as markers of treatment response in Fabry cardiomyopathy. Heart 2016, 102, 1309–1314. [Google Scholar] [CrossRef] [PubMed]
    158. Weidemann, F.; Niemann, M.; Breunig, F.; Herrmann, S.; Beer, M.; Störk, S.; Voelker, W.; Ertl, G.; Wanner, C.; Strotmann, J. Long-term effects of enzyme replacement therapy on Fabry cardiomyopathy: Evidence for a better outcome with early treatment. Circulation 2009, 119, 524–529. [Google Scholar] [CrossRef] [PubMed]
    159. Camporeale, A.; Bandera, F.; Pieroni, M.; Pieruzzi, F.; Spada, M.; Bersano, A.; Econimo, L.; Lanzillo, C.; Rubino, M.; Mignani, R.; et al. Effect of Migalastat on cArdiac InvOlvement in FabRry DiseAse: MAIORA study. J. Med. Genet. 2023, 60, 850–858. [Google Scholar] [CrossRef] [PubMed]
    160. Jovanovic, A.; Schiffmann, R.; Nicholls, K.; Feldt-Rasmussen, U.; Bichet, D.G.; Hughes, D.; Jain, V.; Yu, J.; Castelli, J.; Skuban, N.; et al. Improvements in cardiac mass with long-term migalastat treatment in patients with Fabry disease: Results from phase 3 trials. J. Inborn Errors Metab. Screen. 2017, 5, Abstract LBN 02. [Google Scholar] [CrossRef]
    161. Krämer, J.; Niemann, M.; Störk, S.; Frantz, S.; Beer, M.; Ertl, G.; Wanner, C.; Weidemann, F. Relation of burden of myocardial fibrosis to malignant ventricular arrhythmias and outcomes in Fabry disease. Am. J. Cardiol. 2014, 114, 895–900. [Google Scholar] [CrossRef] [PubMed]
    162. Lee, C.L.; Lin, S.P.; Niu, D.M.; Lin, H.Y. Fabry disease and the effectiveness of enzyme replacement therapy (ERT) in left ventricular hypertrophy (LVH) improvement: A review and meta-analysis. Int. J. Med Sci. 2022, 19, 126–131. [Google Scholar] [CrossRef] [PubMed]
    163. Mignani, R.; Feriozzi, S.; Pisani, A.; Cioni, A.; Comotti, C.; Cossu, M.; Foschi, A.; Giudicissi, A.; Gotti, E.; Lozupone, V.A.; et al. Agalsidase therapy in patients with Fabry disease on renal replacement therapy: A nationwide study in Italy. Nephrol. Dial. Transplant. 2008, 23, 1628–1635. [Google Scholar] [CrossRef] [PubMed]
    164. Figliozzi, S.; Georgiopoulos, G.; Stankowski, K.M.; Camporeale, A.; Pieroni, M. Effects of enzyme replacement therapy on CMR parameters in patients with Fabry disease: A systematic review and meta-analysis. Eur. Heart. J. Suppl. 2022, 24 (Suppl. K), K219–K220. [Google Scholar] [CrossRef]
    165. Pogoda, C.; Brand, S.-M.; Duning, T.; Schmidt-Pogoda, A.; Sindermann, J.; Lenders, M.; Brand, E. Impact of enzyme replacement therapy and migalastat on left atrial strain and cardiomyopathy in patients with Fabry disease. Front. Cardiovasc. Med. 2023, 10, 1223635. [Google Scholar] [CrossRef] [PubMed]
    166. Germain, D.P.; Weidemann, F.; Abiose, A.; Patel, M.R.; Cizmarik, M.; Cole, J.A.; Beitner-Johnson, D.; Benistan, K.; Cabrera, G.; Charrow, J.; et al. Analysis of left ventricular mass in untreated men and in men treated with agalsidase-beta: Data from the Fabry Registry. Genet. Med. 2013, 15, 958–965. [Google Scholar] [CrossRef] [PubMed]
    167. Veldman, B.C.F.; Schoenmakers, D.H.; van Dussen, L.; Datema, M.R.; Langeveld, M. Establishing treatment effectiveness in Fabry disease: Observation-based recommendations for improvement. Int. J. Mol. Sci. 2024, 25, 9752. [Google Scholar] [CrossRef] [PubMed]
    168. Imbriaco, M.; Pisani, A.; Spinelli, L.; Cuocolo, A.; Messalli, G.; Capuano, E.; Marmo, M.; Liuzzi, R.; Visciano, B.; Cianciaruso, B.; et al. Effects of enzyme-replacement therapy in patients with Anderson-Fabry disease: A prospective long-term cardiac magnetic resonance imaging study. Heart 2009, 95, 1103–1107. [Google Scholar] [CrossRef] [PubMed]
    169. Niemann, M.; Breunig, F.; Beer, M.; Herrmann, S.; Strotmann, J.; Hu, K.; Emmert, A.; Voelker, W.; Ertl, G.; Wanner, C.; et al. The right ventricle in Fabry disease: Natural history and impact of enzyme replacement therapy. Heart 2010, 96, 1915–1919. [Google Scholar] [CrossRef] [PubMed]
    170. Weidemann, F.; Breunig, F.; Beer, M.; Knoll, A.; Turschner, O.; Wanner, C.; Sandstede, J.; Voelker, W.; Ertl, G.; Strotmann, J. Improvement of cardiac function during enzyme replacement therapy in patients with Fabry disease: A prospective strain rate imaging study. Circulation 2003, 108, 1299–1301. [Google Scholar] [CrossRef] [PubMed]
    171. Jardim, L.B.; Aesse, F.; Vedolin, L.M.; Pitta-Pinheiro, C.; Marconato, J.; Burin, M.G.; Cecchin, C.; Netto, C.B.; Matte, U.S.; Pereira, F.; et al. White matter lesions in Fabry disease before and after enzyme replacement therapy: A 2-year follow-up. Arq. Neuro-Psiquiatria 2006, 64, 711–717. [Google Scholar] [CrossRef] [PubMed]
    172. Lee, H.-J.; Hsu, T.-R.; Hung, S.-C.; Yu, W.-C.; Chu, T.-H.; Yang, C.-F.; Bizjajeva, S.; Tiu, C.-M.; Niu, D.-M. A comparison of central nervous system involvement in patients with classical Fabry disease or the later-onset subtype with the IVS4 + 919G > A mutation. BMC Neurol. 2017, 17, 25. [Google Scholar] [CrossRef] [PubMed]
    173. Korver, S.; Vergouwe, M.; Hollak, C.E.M.; van Schaik, I.N.; Langeveld, M. Development and clinical consequences of white matter lesions in Fabry disease: A systematic review. Mol. Genet. Metab. 2018, 125, 205–216. [Google Scholar] [CrossRef] [PubMed]
    174. Korver, S.; Longo, M.G.F.; Lima, M.R.; Hollak, C.E.M.; El Sayed, M.; van Schaik, I.N.; Vedolin, L.; Dijkgraaf, M.G.W.; Langeveld, M. Determinants of cerebral radiological progression in Fabry disease. J. Neurol. Neurosurg. Psychiatry 2020, 91, 756–763. [Google Scholar] [CrossRef] [PubMed]
    175. Lulla, D.S.; Higuita, L.; Shukla-Parekh, S.; Lau, H. Survey assessing the prevalence and severity of neuropsychiatric manifestations in patients living with Fabry disease. Mol. Genet. Metab. 2020, 129 (Suppl. S2), S103. [Google Scholar] [CrossRef]
    176. Sunder-Plassmann, G.; Shankar, S.; Wilcox, W.; Nicholls, K.; Giugliani, R.; Lagast, H.; Skuban, N.; Germain, D.P. Occurrence of cerebrovascular events during long-term treatment with migalastat in patients with Fabry disease. J. Inherit. Metab. Dis. 2019, 42 (Suppl. S1), 233. [Google Scholar]
    177. Banikazemi, M.; Bultas, J.; Waldek, S.; Wilcox, W.R.; Whitley, C.B.; McDonald, M.; Finkel, R.; Packman, S.; Bichet, D.G.; Warnock, D.G.; et al. Agalsidase-beta therapy for advanced Fabry disease: A randomized trial. Ann. Intern. Med. 2007, 146, 77–86. [Google Scholar] [CrossRef] [PubMed]
    178. Fellgiebel, A.; Gartenschlager, M.; Wildberger, K.; Scheurich, A.; Desnick, R.J.; Sims, K. Enzyme replacement therapy stabilized white matter lesion progression in Fabry disease. Cerebrovasc. Dis. 2014, 38, 448–456. [Google Scholar] [CrossRef] [PubMed]
    179. Lenders, M.; Karabul, N.; Duning, T.; Schmitz, B.; Schelleckes, M.; Mesters, R.; Hense, H.-W.; Beck, M.; Brand, S.-M.; Brand, E. Thromboembolic events in Fabry disease and the impact of factor v Leiden. Neurology 2015, 84, 1009–1016. [Google Scholar] [CrossRef] [PubMed]
    180. Whybra, C.; Kampmann, C.; Krummenauer, F.; Ries, M.; Mengel, E.; Miebach, E.; Baehner, F.; Kim, K.; Bajbouj, M.; Schwarting, A.; et al. The Mainz Severity Score Index: A new instrument for quantifying the Anderson-Fabry disease phenotype, and the response of patients to enzyme replacement therapy. Clin. Genet. 2004, 65, 299–307. [Google Scholar] [CrossRef] [PubMed]
    181. Parini, R.; Rigoldi, M.; Santus, F.; Furlan, F.; De Lorenzo, P.; Valsecchi, G.; Concolino, D.; Strisciuglio, P.; Feriozzi, S.; Di Vito, R.; et al. Enzyme replacement therapy with agalsidase alfa in a cohort of Italian patients with Anderson-Fabry disease: Testing the effects with the Mainz Severity Score Index. Clin. Genet. 2008, 74, 260–266. [Google Scholar] [CrossRef] [PubMed]
    182. Concolino, D.; Amico, L.; Cappellini, M.D.; Cassinerio, E.; Conti, M.; Donati, M.; Falvo, F.; Fiumara, A.; Maccarone, M.; Manna, R.; et al. Home infusion program with enzyme replacement therapy for Fabry disease: The experience of a large Italian collaborative group. Mol. Genet. Metab. Rep. 2017, 12, 85–91. [Google Scholar] [CrossRef] [PubMed]
    183. Biegstraaten, M.; van Schaik, I.N.; Wieling, W.; Wijburg, F.A.; Hollak, C.E. Autonomic neuropathy in Fabry disease: A prospective study using the Autonomic Symptom Profile and cardiovascular autonomic function tests. BMC Neurol. 2010, 10, 38. [Google Scholar] [CrossRef] [PubMed]
    184. Chen, K.-H.; Chien, Y.; Wang, K.-L.; Leu, H.-B.; Hsiao, C.-Y.; Lai, Y.-H.; Wang, C.-Y.; Chang, Y.-L.; Lin, S.-J.; Niu, D.-M.; et al. Evaluation of proinflammatory prognostic biomarkers for Fabry cardiomyopathy with enzyme replacement therapy. Can. J. Cardiol. 2016, 32, 1221.e1221–1221.e1229. [Google Scholar] [CrossRef] [PubMed]
    185. Neto, N.S.R.; Bento, J.C.B.; Caparbo, V.F.; Pereira, R.M.R. Increased serum interleukin-6 and tumor necrosis factor alpha levels in Fabry disease: Correlation with disease burden. Clinics 2021, 76, e2643. [Google Scholar] [CrossRef] [PubMed]
    186. Chen, K.-H.; Chou, Y.-C.; Hsiao, C.-Y.; Chien, Y.; Wang, K.-L.; Lai, Y.-H.; Chang, Y.-L.; Niu, D.-M.; Yu, W.-C. Amelioration of serum 8-OHdG level by enzyme replacement therapy in patients with Fabry cardiomyopathy. Biochem. Biophys. Res. Commun. 2017, 486, 293–299. [Google Scholar] [CrossRef] [PubMed]
    187. Ripeau, D.; Amartino, H.; Cedrolla, M.; Urtiaga, L.; Urdaneta, B.; Cano, M.; Valdez, R.; Antongiovanni, N.; Masllorens, F. Cambio de agalsidasa beta por agalsidasa alfa en la terapia de reemplazo enzimatica de pacientes con enfermedad de Fabry en Latinoamerica. Medicina 2017, 77, 173–179. [Google Scholar] [PubMed]
    188. Orsborne, C.; Black, N.; Naish, J.H.; Woolfson, P.; Reid, A.B.; Schmitt, M.; Jovanovic, A.; A Miller, C. Disease-specific therapy for the treatment of the cardiovascular manifestations of Fabry disease: A systematic review. Heart 2023, 110, 19–26. [Google Scholar] [CrossRef] [PubMed]
    189. Besekar, S.M.; Jogdand, S.D.; Naqvi, W.M. Fabry disease and its management: A literature analysis. Cureus 2023, 15, e37048. [Google Scholar] [CrossRef] [PubMed]
    190. Riccio, E.; Pisani, A. New insights in efficacy of different enzyme replacement therapy dosages in Fabry disease: Switch studies data following agalsidase beta shortage. Clin. Genet. 2023, 103, 371–376. [Google Scholar] [CrossRef] [PubMed]
    191. Riccio, E.; de Marco, O.; Pisani, A. Medication adherence in Fabry patients treated with migalastat: Real world experience. Mol. Genet. Metab. Rep. 2023, 35, 100976. [Google Scholar] [CrossRef] [PubMed]
    192. Ramaswami, U.; Pintos-Morell, G.; Kampmann, C.; Nicholls, K.; Niu, D.-M.; Reisin, R.; West, M.L.; Anagnostopoulou, C.; Botha, J.; Jazukeviciene, D.; et al. Two decades of experience of the Fabry Outcome Survey provides further confirmation of the long-term effectiveness of agalsidase alfa enzyme replacement therapy. Mol. Genet. Metab. Rep. 2025, 43, 101215. [Google Scholar] [CrossRef] [PubMed]
    193. Khanna, R.; Soska, R.; Lun, Y.; Feng, J.; Frascella, M.; Young, B.; Brignol, N.; Pellegrino, L.; A Sitaraman, S.; Desnick, R.J.; et al. The pharmacological chaperone 1-deoxygalactonojirimycin reduces tissue globotriaosylceramide levels in a mouse model of Fabry disease. Mol. Ther. 2010, 18, 23–33. [Google Scholar] [CrossRef] [PubMed]
    194. Burlina, A.; Al-Shaar, L.; Germain, D.P.; Banikazemi, M.; Ponce, E.; Crespo, A.; Lawson, V.; Politei, J. Reduced incidence of stroke in patients with Fabry disease treated with agalsidase beta: A matched analysis from the Fabry Registry. Mol. Genet. Metab. 2025, 144, 16. [Google Scholar] [CrossRef]
    195. Feriozzi, S.; Chimenti, C.; Reisin, R.C. Updated evaluation of agalsidase alfa enzyme replacement therapy for patients with Fabry disease: Insights from real-world data. Drug Des. Dev. Ther. 2024, 18, 1083–1101. [Google Scholar] [CrossRef] [PubMed]
    196. Batista, J.L.; Hariri, A.; Maski, M.; Richards, S.; Gudivada, B.; A Raynor, L.; Ponce, E.; Wanner, C.; Desnick, R.J. Reduction in kidney function decline and risk of severe clinical events in agalsidase beta-treated Fabry disease patients: A matched analysis from the Fabry Registry. Clin. Kidney J. 2024, 17, sfae194. [Google Scholar] [CrossRef] [PubMed]
    197. Pisani, A.; Wilson, K.M.; Batista, J.L.; Kantola, I.; Ortiz, A.; Politei, J.; Al-Shaar, L.; Maski, M.; Crespo, A.; Ponce, E.; et al. Clinical outcomes in patients switching from agalsidase beta to migalastat: A Fabry Registry analysis. J. Inherit. Metab. Dis. 2024, 47, 1080–1095. [Google Scholar] [CrossRef] [PubMed]
    198. Lenders, M.; Nowak, A.; Cybulla, M.; Kaufeld, J.; Köhn, A.F.; Muschol, N.M.; Kurschat, C.; Brand, E. Impact of enzyme replacement therapy and migalastat on disease progression in females with Fabry disease. Orphanet J. Rare Dis. 2025, 20, 79. [Google Scholar] [CrossRef] [PubMed]
    199. Hughes, D.A.; Sunder-Plassmann, G.; Jovanovic, A.; Brand, E.; West, M.L.; Bichet, D.G.; Pisani, A.; Nowak, A.; Torra, R.; Khan, A.; et al. Renal and multisystem effectiveness of 3.9 years of migalastat in a global real-world cohort: Results from the followME Fabry Pathfinders registry. J. Inherit. Metab. Dis. 2025, 48, e12771. [Google Scholar] [CrossRef] [PubMed]
    200. Ramaswami, U.; Font-Montgomery, E.; Goker-Alpan, O.; Ortiz, D.; Sanchez-Valle, A.; Whitley, C.B.; Wilcox, W.R.; Jiang, H.; Lawson, L.A.; Vosk, J.; et al. Safety and efficacy of migalastat in adolescent patients with Fabry disease: Results from ASPIRE, a phase 3b, open-label, single-arm, 12-month clinical trial, and its open-label extension. Mol. Genet. Metab. 2025, 145, 109102. [Google Scholar] [CrossRef] [PubMed]
    201. Nowak, A.; Botha, J.; Anagnostopoulou, C.; Hughes, D.A. Clinical outcomes in elderly patients receiving agalsidase alfa treatment in the Fabry Outcome Survey. Mol. Genet. Metab. 2024, 143, 108561. [Google Scholar] [CrossRef] [PubMed]
    202. Arakawa, M.; Ikeda, Y.; Otaka, H.; Iwashiro, S. Long-term safety of enzyme replacement therapy with agalsidase alfa in patients with Fabry disease: Post-marketing extension surveillance in Japan. Mol. Genet. Metab. Rep. 2024, 40, 101122. [Google Scholar] [CrossRef] [PubMed]
    203. Liu, Y.; Li, Y.; Li, P.; Zhang, S.; Zhiqing, Z. Effectiveness and safety of enzyme replacement therapy in the treatment of Fabry disease: A Chinese monocentric real-world study. Orphanet J. Rare Dis. 2024, 19, 422. [Google Scholar] [CrossRef] [PubMed]
    204. Holida, M.; Linhart, A.; Pisani, A.; Longo, N.; Eyskens, F.; Goker-Alpan, O.; Wallace, E.; Deegan, P.; Tøndel, C.; Feldt-Rasmussen, U.; et al. A phase III, open-label clinical trial evaluating pegunigalsidase alfa administered every 4 weeks in adults with Fabry disease previously treated with other enzyme replacement therapies. J. Inherit. Metab. Dis. 2025, 48, e12795. [Google Scholar] [CrossRef] [PubMed]
    Figure 1. PRISMA flow diagram for identified publications (initial and updated searches). PRISMA, Preferred Reporting Items for Systematic reviews and Meta-Analyses.
    Figure 1. PRISMA flow diagram for identified publications (initial and updated searches). PRISMA, Preferred Reporting Items for Systematic reviews and Meta-Analyses.
    Jcm 14 05131 g001
    Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

    Share and Cite

    MDPI and ACS Style

    Jovanovic, A.; Miller-Hodges, E.; Castriota, F.; Evuarherhe, O.; Ayodele, O.; Hughes, D.; Pintos-Morell, G.; Giugliani, R.; Feriozzi, S.; Siffel, C. Clinical Efficacy and Real-World Effectiveness of Fabry Disease Treatments: A Systematic Literature Review. J. Clin. Med. 2025, 14, 5131. https://doi.org/10.3390/jcm14145131

    AMA Style

    Jovanovic A, Miller-Hodges E, Castriota F, Evuarherhe O, Ayodele O, Hughes D, Pintos-Morell G, Giugliani R, Feriozzi S, Siffel C. Clinical Efficacy and Real-World Effectiveness of Fabry Disease Treatments: A Systematic Literature Review. Journal of Clinical Medicine. 2025; 14(14):5131. https://doi.org/10.3390/jcm14145131

    Chicago/Turabian Style

    Jovanovic, Ana, Eve Miller-Hodges, Felicia Castriota, Obaro Evuarherhe, Olulade Ayodele, Derralynn Hughes, Guillem Pintos-Morell, Roberto Giugliani, Sandro Feriozzi, and Csaba Siffel. 2025. "Clinical Efficacy and Real-World Effectiveness of Fabry Disease Treatments: A Systematic Literature Review" Journal of Clinical Medicine 14, no. 14: 5131. https://doi.org/10.3390/jcm14145131

    APA Style

    Jovanovic, A., Miller-Hodges, E., Castriota, F., Evuarherhe, O., Ayodele, O., Hughes, D., Pintos-Morell, G., Giugliani, R., Feriozzi, S., & Siffel, C. (2025). Clinical Efficacy and Real-World Effectiveness of Fabry Disease Treatments: A Systematic Literature Review. Journal of Clinical Medicine, 14(14), 5131. https://doi.org/10.3390/jcm14145131

    Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

    Article Metrics

    Back to TopTop