1
|
Berstein V, Pirotzky EM, Taconelli HD, Gobbi MG, Beider L, Salgueiro ND, Dome L, Diez RA, Sotelo H, Coppola S. Comparative pharmacokinetics and pharmacodynamics of two formulations of agalsidase beta (agalsidase Biosidus) and Fabrazyme® by intravenous infusion in healthy male volunteers. Mol Genet Metab Rep 2024; 41:101149. [PMID: 39435314 PMCID: PMC11492607 DOI: 10.1016/j.ymgmr.2024.101149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/23/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024] Open
Abstract
Fabry disease is a rare X-linked lysosomal condition that leads to the accumulation of glycosphingolipids in various tissues, causing cellular dysfunction, tissue remodeling, progressive fibrosis, and organ failure. The disease results from a deficiency in the human α-galactosidase A enzyme, responsible for breaking down glycosphingolipids like globotriaosylceramide (GL-3 or Gb3) into galactose and dihexose ceramides. In individuals diagnosed with Fabry disease, treatment from 2 years of age onwards typically involves agalsidase beta, the normal recombinant form of the defective enzyme. Agalsidase beta from Biosidus has been developed as a biosimilar to Sanofi-Genzyme's Fabrazyme®. In the molecule's clinical journey, a phase I trial was designed to establish its similarity in terms of pharmacokinetics, pharmacodynamics, and immunogenicity compared to the reference medication. The study was conducted on 24 healthy male volunteers, aged between 18 and 40 years. All volunteers received a single 1 mg/kg bw dose of Fabrazyme® or Biosidus Agalsidase beta by continuous intravenous (IV) infusion over 5 h. The 90 % confidence interval (CI) of the maximum concentration (Cmax), area under the plasma concentration-time curve from time 0 to 12 h (AUC0-12 h) and area under the plasma concentration-time curve extrapolated from time 0 to infinity (AUC0-∞) ratios fell within the accepted range of 80-125 %. No differences were detected in adverse effects or antibody induction. This indicates that Biosidus agalsidase beta meets the criteria for being considered similar to the reference formulation Sanofi Genzyme's Fabrazyme®.
Collapse
Affiliation(s)
| | | | | | | | - Lara Beider
- Biosidus S.A.U, Buenos Aires 1254, Argentina
| | | | - Laila Dome
- Biosidus S.A.U, Buenos Aires 1254, Argentina
| | | | - Hugo Sotelo
- Biosidus S.A.U, Buenos Aires 1254, Argentina
| | | |
Collapse
|
2
|
Holida M, Linhart A, Pisani A, Longo N, Eyskens F, Goker-Alpan O, Wallace E, Deegan P, Tøndel C, Feldt-Rasmussen U, Hughes D, Sakov A, Rocco R, Almon EB, Alon S, Chertkoff R, Warnock DG, Waldek S, Wilcox WR, Bernat JA. 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 2024. [PMID: 39381863 DOI: 10.1002/jimd.12795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 10/10/2024]
Abstract
Pegunigalsidase alfa, a PEGylated α-galactosidase A enzyme replacement therapy (ERT) for Fabry disease, has a longer plasma half-life than other ERTs administered intravenously every 2 weeks (E2W). BRIGHT (NCT03180840) was a phase III, open-label study in adults with Fabry disease, previously treated with agalsidase alfa or beta E2W for ≥3 years, who switched to 2 mg/kg pegunigalsidase alfa every 4 weeks (E4W) for 52 weeks. Primary objective assessed safety, including number of treatment-emergent adverse events (TEAEs). Thirty patients were enrolled (24 males); 23 previously received agalsidase beta. Pegunigalsidase alfa plasma concentrations remained above the lower limit of quantification throughout the 4-week dosing interval. Thirty-three of 182 TEAEs (in 9 patients) were considered treatment-related; all were mild/moderate. No patients developed de novo anti-drug antibodies (ADAs). In the efficacy analysis (n = 29), median (inter-quartile range) eGFR change from baseline over 52 weeks was -1.9 (-5.9; 1.8) mL/min/1.73 m2 (n = 28; males [n = 22]: -2.4 [-5.2; 3.2]; females [n = 6]: -0.7 [-9.2; 2.0]). Overall, median eGFR slope was -1.9 (-8.3; 1.9) mL/min/1.73 m2/year (ADA-negative [n = 20]: -1.2 [-6.4; 2.6]; ADA-positive [n = 9]: -8.4 [-11.6; -1.0]). Lyso-Gb3 concentrations were low and stable in females, with a slight increase in males (9/24 ADA-positive). The BRIGHT study results suggest that 2 mg/kg pegunigalsidase alfa E4W is tolerated well in stable adult patients with Fabry disease. Due to the low number of patients in this study, more research is needed to demonstrate the effects of pegunigalsidase alfa given E4W. Further evidence, outside of this clinical trial, should be factored in for physicians to prolong the biweekly ERT intervals to E4W. TAKE-HOME MESSAGE: Treatment with 2 mg/kg pegunigalsidase alfa every 4 weeks could offer a new treatment option for patients with Fabry disease.
Collapse
Affiliation(s)
- Myrl Holida
- Division of Medical Genetics and Genomics, Stead Family Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Aleš Linhart
- Charles University, General University Hospital, Prague, Czech Republic
| | - Antonio Pisani
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Nicola Longo
- Pediatrics Medical Genetics, University of Utah, Salt Lake City, Utah, USA
| | | | - Ozlem Goker-Alpan
- Lysosomal and Rare Disorders Research and Treatment Center, Fairfax, Virginia, USA
| | - Eric Wallace
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Patrick Deegan
- Lysosomal Disorders Unit, Cambridge University Hospitals NHS Foundation Trust and University of Cambridge, Cambridge, UK
| | - Camilla Tøndel
- University of Bergen and Haukeland University Hospital, Bergen, Norway
| | - Ulla Feldt-Rasmussen
- Department of Endocrinology and Metabolism, Rigshospitalet and Faculty of Health and Clinical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Derralynn Hughes
- LSDU, Royal Free London NHS Foundation Trust, and University College London, London, UK
| | | | | | - Einat Brill Almon
- Department of Product Development, Protalix Biotherapeutics, Carmiel, Israel
| | - Sari Alon
- Department of Product Development, Protalix Biotherapeutics, Carmiel, Israel
| | - Raul Chertkoff
- Department of Product Development, Protalix Biotherapeutics, Carmiel, Israel
| | - David G Warnock
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - William R Wilcox
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - John A Bernat
- Division of Medical Genetics and Genomics, Stead Family Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
3
|
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. [PMID: 39077747 PMCID: PMC11284380 DOI: 10.1016/j.ymgmr.2024.101122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/31/2024] Open
Abstract
Fabry disease is a rare inherited X-linked metabolic disorder in which deficient alpha-galactosidase A activity causes progressive build-up of globotriaosylceramide (Gb3) and multi-system dysfunction. Following approval of agalsidase alfa for Fabry disease in Japan in 2006, an 8-year all-case post-marketing surveillance (PMS) showed that the treatment was well tolerated and effective for managing disease progression in adult Japanese patients. The present nationwide prospective observational study extended the initial PMS by enrolling patients who continued agalsidase alfa treatment after the initial 8-year period in a 6.5-year extension survey. Patient information from the initial PMS and the extension survey was evaluated as a single data set (observation period: February 2007-September 2021). Of 493 patients in the initial PMS, 129 (45.0% male classic, 6.2% male non-classic, 48.8% female heterozygous phenotype) consented to participate in the extension survey and were included in the analysis. The mean duration of treatment was 9.6 years. A total of 145 adverse drug reactions (ADRs) occurred in 31 patients (24%), and 22 serious ADRs occurred in 12 patients (9.3%). Although serious cardiac, renal, or cerebrovascular adverse events decreased in frequency over time in male patients, serious cardiac events continued to occur in female patients, who showed higher incidence of cardiac complications at baseline. No new safety concerns were identified. Additionally, long-term agalsidase alfa treatment sustained the initial reduction in Gb3 concentrations without increasing the rate of anti-agalsidase antibody positivity. These findings suggest that agalsidase alfa treatment demonstrates continued safety and sustains patients' clinical course over the long term.
Collapse
Affiliation(s)
- Makoto Arakawa
- Japan Medical Office, Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Yoshinori Ikeda
- Japan Medical Office, Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Hiromichi Otaka
- Japan Medical Office, Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Sanghun Iwashiro
- Japan Medical Office, Takeda Pharmaceutical Company Limited, Tokyo, Japan
| |
Collapse
|
4
|
Wallace EL, Goker-Alpan O, Wilcox WR, Holida M, Bernat J, Longo N, Linhart A, Hughes DA, Hopkin RJ, Tøndel C, Langeveld M, Giraldo P, Pisani A, Germain DP, Mehta A, Deegan PB, Molnar MJ, Ortiz D, Jovanovic A, Muriello M, Barshop BA, Kimonis V, Vujkovac B, Nowak A, Geberhiwot T, Kantola I, Knoll J, Waldek S, Nedd K, Karaa A, Brill-Almon E, Alon S, Chertkoff R, Rocco R, Sakov A, Warnock DG. 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 2024; 61:520-530. [PMID: 37940383 PMCID: PMC11137442 DOI: 10.1136/jmg-2023-109445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/10/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Pegunigalsidase alfa is a PEGylated α-galactosidase A enzyme replacement therapy. BALANCE (NCT02795676) assessed non-inferiority of pegunigalsidase alfa versus agalsidase beta in adults with Fabry disease with an annualised estimated glomerular filtration rate (eGFR) slope more negative than -2 mL/min/1.73 m2/year who had received agalsidase beta for ≥1 year. METHODS Patients were randomly assigned 2:1 to receive 1 mg/kg pegunigalsidase alfa or agalsidase beta every 2 weeks for 2 years. The primary efficacy analysis assessed non-inferiority based on median annualised eGFR slope differences between treatment arms. RESULTS Seventy-seven patients received either pegunigalsidase alfa (n=52) or agalsidase beta (n=25). At baseline, mean (range) age was 44 (18-60) years, 47 (61%) patients were male, median eGFR was 74.5 mL/min/1.73 m2 and median (range) eGFR slope was -7.3 (-30.5, 6.3) mL/min/1.73 m2/year. At 2 years, the difference between median eGFR slopes was -0.36 mL/min/1.73 m2/year, meeting the prespecified non-inferiority margin. Minimal changes were observed in lyso-Gb3 concentrations in both treatment arms at 2 years. Proportions of patients experiencing treatment-related adverse events and mild or moderate infusion-related reactions were similar in both groups, yet exposure-adjusted rates were 3.6-fold and 7.8-fold higher, respectively, with agalsidase beta than pegunigalsidase alfa. At the end of the study, neutralising antibodies were detected in 7 out of 47 (15%) pegunigalsidase alfa-treated patients and 6 out of 23 (26%) agalsidase beta-treated patients. There were no deaths. CONCLUSIONS Based on rate of eGFR decline over 2 years, pegunigalsidase alfa was non-inferior to agalsidase beta. Pegunigalsidase alfa had lower rates of treatment-emergent adverse events and mild or moderate infusion-related reactions. TRIAL REGISTRATION NUMBER NCT02795676.
Collapse
Affiliation(s)
- Eric L Wallace
- Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ozlem Goker-Alpan
- Lysosomal and Rare Disorders Research and Treatment Center, Inc, Fairfax, Virginia, USA
| | - William R Wilcox
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Myrl Holida
- Department of Pediatrics, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - John Bernat
- Department of Pediatrics, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Nicola Longo
- Department of Pediatrics, Division of Medical Genetics, University of Utah Health, Salt Lake City, Utah, USA
| | - Aleš Linhart
- Department of Internal Medicine, School of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Derralynn A Hughes
- Lysosomal Storage Disorders Unit, Royal Free London NHS Foundation Trust and University College London, London, UK
| | - Robert J Hopkin
- Department of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Camilla Tøndel
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Mirjam Langeveld
- Academisch Medisch Centrum Universiteit van Amsterdam, Amsterdam, The Netherlands
| | - Pilar Giraldo
- Unidad de Investigación Traslacional. Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), Zaragoza, Spain
| | - Antonio Pisani
- Department of Public Health, Universita degli Studi di Napoli Federico II, Napoli, Italy
| | | | - Ankit Mehta
- Baylor University Medical Center at Dallas, Dallas, Texas, USA
| | - Patrick B Deegan
- Lysosmal Disorders Unit, Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Maria Judit Molnar
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University Clinical Center, Budapest, Hungary
| | - Damara Ortiz
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ana Jovanovic
- Mark Holland Metabolic Unit, Northern Care Alliance NHS Foundation Trust, Greater Manchester, UK
| | - Michael Muriello
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Bruce A Barshop
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Virginia Kimonis
- Department of Pediatrics, University of California Irvine, Irvine, California, USA
| | - Bojan Vujkovac
- Department of Internal Medicine, General Hospital Slovenj Gradec, Slovenj Gradec, Slovenia
| | - Albina Nowak
- Department of Endocrinology and Clinical Nutrition, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Tarekegn Geberhiwot
- Department of Diabetes, Endocrinology and Metabolism, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Ilkka Kantola
- Division of Medicine, Turku University Hospital, Turku, Finland
| | | | | | - Khan Nedd
- Infusion Associates, Grand Rapids, Michigan, USA
| | - Amel Karaa
- Massachusetts General Hospital for Children, Boston, Massachusetts, USA
| | | | - Sari Alon
- Product Development, Protalix Biotherapeutics, Carmiel, Israel
| | | | | | | | - David G Warnock
- Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
5
|
Linhart A, Dostálová G, Nicholls K, West ML, Tøndel C, Jovanovic A, Giraldo P, Vujkovac B, Geberhiwot T, Brill-Almon E, Alon S, Chertkoff R, Rocco R, Hughes D. 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. [PMID: 37865771 PMCID: PMC10589982 DOI: 10.1186/s13023-023-02937-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 09/27/2023] [Indexed: 10/23/2023] Open
Abstract
BACKGROUND Pegunigalsidase alfa is a novel, PEGylated α-galactosidase-A enzyme-replacement therapy approved in the EU and US to treat patients with Fabry disease (FD). OBJECTIVE/METHODS BRIDGE is a phase 3 open-label, switch-over study designed to assess safety and efficacy of 12 months of pegunigalsidase alfa (1 mg/kg every 2 weeks) treatment in adults with FD who had been previously treated with agalsidase alfa (0.2 mg/kg every 2 weeks) for ≥ 2 years. RESULTS Twenty-seven patients were screened; 22 met eligibility criteria; and 20 (13 men, 7 women) completed the study. Pegunigalsidase alfa was well-tolerated, with 97% of treatment-emergent adverse events (TEAEs) being of mild or moderate severity. The incidence of treatment-related TEAEs was low, with 2 (9%) discontinuations due to TEAEs. Five patients (23%) reported infusion-related reactions. Overall mean (SD; n = 22) baseline estimated glomerular filtration rate (eGFR) was 82.5 (23.4) mL/min/1.73 m2 and plasma lyso-Gb3 level was 38.3 (41.2) nmol/L (men: 49.7 [45.8] nmol/L; women: 13.8 [6.1] nmol/L). Before switching to pegunigalsidase alfa, mean (standard error [SE]) annualized eGFR slope was - 5.90 (1.34) mL/min/1.73 m2/year; 12 months post-switch, the mean eGFR slope was - 1.19 (1.77) mL/min/1.73 m2/year; and mean plasma lyso-Gb3 reduced by 31%. Seven (35%) out of 20 patients were positive for pegunigalsidase alfa antidrug antibodies (ADAs) at ≥ 1 study timepoint, two of whom had pre-existing ADAs at baseline. Mean (SE) changes in eGFR slope for ADA-positive and ADA-negative patients were + 5.47 (3.03) and + 4.29 (3.15) mL/min/1.73 m2/year, respectively, suggesting no negative impact of anti-pegunigalsidase alfa ADAs on eGFR slope. CONCLUSION Pegunigalsidase alfa may offer a safe and effective treatment option for patients with FD, including those previously treated with agalsidase alfa. TRN: NCT03018730. Date of registration: January 2017.
Collapse
Affiliation(s)
- Aleš Linhart
- 2nd Department of Internal Cardiovascular Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08, Prague 2, Czech Republic.
| | - Gabriela Dostálová
- 2nd Department of Internal Cardiovascular Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08, Prague 2, Czech Republic
| | - Kathy Nicholls
- Department of Nephrology, Royal Melbourne Hospital and The University of Melbourne, Parkville, Australia
| | - Michael L West
- Division of Nephrology, Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Camilla Tøndel
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Nephrology and Rheumatology Unit, Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Ana Jovanovic
- Department of Inherited Metabolic Disease, Salford Royal, Salford, England, UK
| | - Pilar Giraldo
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Hospital de Dia Quiron, Zaragoza, Spain
| | - Bojan Vujkovac
- Department of Internal Medicine, General Hospital Slovenj Gradec, Slovenj Gradec, Slovenia
| | - Tarekegn Geberhiwot
- Department of Diabetes, Endocrinology and Metabolism, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, England, UK
| | | | - Sari Alon
- Protalix Biotherapeutics, Carmiel, Israel
| | | | | | - Derralynn Hughes
- Lysosomal Storage Disorders Unit, Royal Free London NHS Foundation Trust and University College London, London, England, UK
| |
Collapse
|
6
|
Averbuch T, White JA, Fine NM. Anderson-Fabry disease cardiomyopathy: an update on epidemiology, diagnostic approach, management and monitoring strategies. Front Cardiovasc Med 2023; 10:1152568. [PMID: 37332587 PMCID: PMC10272370 DOI: 10.3389/fcvm.2023.1152568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/09/2023] [Indexed: 06/20/2023] Open
Abstract
Anderson-Fabry disease (AFD) is an X-linked lysosomal storage disorder caused by deficient activity of the enzyme alpha-galactosidase. While AFD is recognized as a progressive multi-system disorder, infiltrative cardiomyopathy causing a number of cardiovascular manifestations is recognized as an important complication of this disease. AFD affects both men and women, although the clinical presentation typically varies by sex, with men presenting at a younger age with more neurologic and renal phenotype and women developing a later onset variant with more cardiovascular manifestations. AFD is an important cause of increased myocardial wall thickness, and advances in imaging, in particular cardiac magnetic resonance imaging and T1 mapping techniques, have improved the ability to identify this disease non-invasively. Diagnosis is confirmed by the presence of low alpha-galactosidase activity and identification of a mutation in the GLA gene. Enzyme replacement therapy remains the mainstay of disease modifying therapy, with two formulations currently approved. In addition, newer treatments such as oral chaperone therapy are now available for select patients, with a number of other investigational therapies in development. The availability of these therapies has significantly improved outcomes for AFD patients. Improved survival and the availability of multiple agents has presented new clinical dilemmas regarding disease monitoring and surveillance using clinical, imaging and laboratory biomarkers, in addition to improved approaches to managing cardiovascular risk factors and AFD complications. This review will provide an update on clinical recognition and diagnostic approaches including differentiation from other causes of increased ventricular wall thickness, in addition to modern strategies for management and follow-up.
Collapse
Affiliation(s)
- Tauben Averbuch
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
| | - James A. White
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
- Stephenson Cardiac Imaging Center, Alberta Health Services, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nowell M. Fine
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
7
|
Burlina A, Brand E, Hughes D, Kantola I, Krӓmer J, Nowak A, Tøndel C, Wanner C, Spada M. An expert consensus on the recommendations for the use of biomarkers in Fabry disease. Mol Genet Metab 2023; 139:107585. [PMID: 37207471 DOI: 10.1016/j.ymgme.2023.107585] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
Fabry disease is an X-linked lysosomal storage disorder caused by the accumulation of glycosphingolipids in various tissues and body fluids, leading to progressive organ damage and life-threatening complications. Phenotypic classification is based on disease progression and severity and can be used to predict outcomes. Patients with a classic Fabry phenotype have little to no residual α-Gal A activity and have widespread organ involvement, whereas patients with a later-onset phenotype have residual α-Gal A activity and disease progression can be limited to a single organ, often the heart. Diagnosis and monitoring of patients with Fabry disease should therefore be individualized, and biomarkers are available to support with this. Disease-specific biomarkers are useful in the diagnosis of Fabry disease; non-disease-specific biomarkers may be useful to assess organ damage. For most biomarkers it can be challenging to prove they translate to differences in the risk of clinical events associated with Fabry disease. Therefore, careful monitoring of treatment outcomes and collection of prospective data in patients are needed. As we deepen our understanding of Fabry disease, it is important to regularly re-evaluate and appraise published evidence relating to biomarkers. In this article, we present the results of a literature review of evidence published between February 2017 and July 2020 on the impact of disease-specific treatment on biomarkers and provide an expert consensus on clinical recommendations for the use of those biomarkers.
Collapse
Affiliation(s)
- Alessandro Burlina
- Neurological Unit, St. Bassiano Hospital, Via dei Lotti 40, I-36061 Bassano del Grappa, Italy.
| | - Eva Brand
- Internal Medicine, Department of Nephrology, Hypertension and Rheumatology; Interdisciplinary Fabry Center Münster (IFAZ), University Hospital Münster, Münster, Germany
| | - Derralynn Hughes
- Lysosomal Storage Disorders Unit, Royal Free London NHS Foundation Trust, University College London, United Kingdom
| | - Ilkka Kantola
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Johannes Krӓmer
- Pediatric Neurology and Metabolism, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Albina Nowak
- Department of Endocrinology and Clinical Nutrition, University Hospital of Zurich, Zurich, Switzerland
| | - Camilla Tøndel
- Department of Clinical Science, University of Bergen and Department of Paediatrics, Haukeland University Hospital, Bergen, Norway
| | - Christoph Wanner
- Department of Internal Medicine, Division of Nephrology, Fabry Center for Interdisciplinary Therapy (FAZIT), University Hospital of Würzburg, Würzburg, Germany
| | - Marco Spada
- Department of Pediatrics, University of Torino, Torino, Italy
| |
Collapse
|
8
|
Antibodies against recombinant enzyme in the treatment of Fabry disease: Now you see them, now you don’t. MOLECULAR THERAPY - METHODS & CLINICAL DEVELOPMENT 2022; 27:324-326. [DOI: 10.1016/j.omtm.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
9
|
Silva CAB, Andrade LGMD, Vaisbich MH, Barreto FDC. Brazilian consensus recommendations for the diagnosis, screening, and treatment of individuals with fabry disease: Committee for Rare Diseases - Brazilian Society of Nephrology/2021. J Bras Nefrol 2022; 44:249-267. [PMID: 35212703 PMCID: PMC9269181 DOI: 10.1590/2175-8239-jbn-2021-0208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/16/2021] [Indexed: 11/21/2022] Open
Abstract
Fabry disease (FD) is an X-linked inherited disorder caused by mutations in the GLA gene encoding enzyme alpha-galactosidase A (α-Gal A). The purpose of this study was to produce a consensus statement to standardize the recommendations concerning kidney involvement in FD and provide advice on the diagnosis, screening, and treatment of adult and pediatric patients. This consensus document was organized from an initiative led by the Committee for Rare Diseases (Comdora) of the Brazilian Society of Nephrology (SBN). The review considered randomized clinical trials, real-world data studies, and the expertise of its authors. The purpose of this consensus statement is to help manage patient and physician expectations concerning the outcomes of treatment. Our recommendations must be interpreted within the context of available evidence. The decisions pertaining to each individual case must be made with the involvement of patients and their families and take into account not only the potential cost of treatment, but also concurrent conditions and personal preferences. The Comdora intends to update these recommendations regularly so as to reflect recent literature evidence, real-world data, and appreciate the professional experience of those involved. This consensus document establishes clear criteria for the diagnosis of FD and for when to start or stop specific therapies or adjuvant measures, to thus advise the medical community and standardize clinical practice.
Collapse
|
10
|
Lenders M, Brand E. Mechanisms of Neutralizing Anti-drug Antibody Formation and Clinical Relevance on Therapeutic Efficacy of Enzyme Replacement Therapies in Fabry Disease. Drugs 2021; 81:1969-1981. [PMID: 34748189 PMCID: PMC8602155 DOI: 10.1007/s40265-021-01621-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2021] [Indexed: 12/13/2022]
Abstract
Fabry disease (FD) is a rare X-linked lysosomal storage disorder caused by mutations in the α-galactosidase A (AGAL/GLA) gene. The lysosomal accumulation of the substrates globotriaosylceramide (Gb3) and globotriaosylsphingosine (lyso-Gb3) results in progressive renal failure, cardiomyopathy associated with cardiac arrhythmia, and recurrent strokes, significantly limiting life expectancy in affected patients. Current treatment options for FD include recombinant enzyme-replacement therapies (ERTs) with intravenous agalsidase-α (0.2 mg/kg body weight) or agalsidase-β (1 mg/kg body weight) every 2 weeks, facilitating cellular Gb3 clearance and an overall improvement of disease burden. However, ERT can lead to infusion-associated reactions, as well as the formation of neutralizing anti-drug antibodies (ADAs) in ERT-treated males, leading to an attenuation of therapy efficacy and thus disease progression. In this narrative review, we provide a brief overview of the clinical picture of FD and diagnostic confirmation. The focus is on the biochemical and clinical significance of neutralizing ADAs as a humoral response to ERT. In addition, we provide an overview of different methods for ADA measurement and characterization, as well as potential therapeutic approaches to prevent or eliminate ADAs in affected patients, which is representative for other ERT-treated lysosomal storage diseases.
Collapse
Affiliation(s)
- Malte Lenders
- Department of Internal Medicine D, Nephrology, Hypertension and Rheumatology, Interdisciplinary Fabry Center Münster (IFAZ), University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany.
| | - Eva Brand
- Department of Internal Medicine D, Nephrology, Hypertension and Rheumatology, Interdisciplinary Fabry Center Münster (IFAZ), University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany
| |
Collapse
|