Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Pol Boudes is active.

Publication


Featured researches published by Pol Boudes.


Orphanet Journal of Rare Diseases | 2012

Safety and pharmacodynamic effects of a pharmacological chaperone on α-galactosidase A activity and globotriaosylceramide clearance in Fabry disease: report from two phase 2 clinical studies

Dominique P. Germain; Roberto Giugliani; Derralynn Hughes; Atul Mehta; Kathy Nicholls; Laura Barisoni; Charles Jennette; Alexander Bragat; Jeff Castelli; Sheela Sitaraman; David J. Lockhart; Pol Boudes

BackgroundFabry disease (FD) is a genetic disorder resulting from deficiency of the lysosomal enzyme α-galactosidase A (α-Gal A), which leads to globotriaosylceramide (GL-3) accumulation in multiple tissues. We report on the safety and pharmacodynamics of migalastat hydrochloride, an investigational pharmacological chaperone given orally at 150 mg every-other-day.MethodsTwo open-label uncontrolled phase 2 studies of 12 and 24 weeks (NCT00283959 and NCT00283933) in 9 males with FD were combined. At multiple time points, α-Gal A activity and GL-3 levels were quantified in blood cells, kidney and skin. GL-3 levels were also evaluated through skin and renal histology.ResultsCompared to baseline, increased α-Gal A activity of at least 50% was demonstrated in blood, skin and kidney in 6 of 9 patients. Patients’ increased α-Gal A activities paralleled the α-Gal A increases observed in vitro in HEK-293 cells transfected with the corresponding mutant form of the enzyme. The same 6 patients who demonstrated increases of α-Gal A activity also had GL-3 reduction in skin, urine and/or kidney, and had α-Gal A mutations that responded in transfected cells incubated with the drug. The 3 patients who did not show a consistent response in vivo had α-Gal A mutations that did not respond to migalastat HCl in transfected cells. Migalastat HCl was well tolerated.ConclusionsMigalastat HCl is a candidate pharmacological chaperone that provides a novel genotype-specific treatment for FD. It enhanced α-Gal A activity and resulted in GL-3 substrate decrease in patients with responsive GLA mutations. Phase 3 studies are ongoing.Trial registrationClinicaltrial.gov: NCT00283959 and NCT00283933


Human Mutation | 2011

A pharmacogenetic approach to identify mutant forms of α‐galactosidase a that respond to a pharmacological chaperone for Fabry disease

Xiaoyang Wu; Evan Katz; Maria Cecilia Della Valle; Kirsten Mascioli; John J. Flanagan; Jeffrey P. Castelli; Raphael Schiffmann; Pol Boudes; David J. Lockhart; Kenneth J. Valenzano; Elfrida R. Benjamin

Fabry disease is caused by mutations in the gene (GLA) that encodes α‐galactosidase A (α‐Gal A). The iminosugar AT1001 (GR181413A, migalastat hydrochloride, 1‐deoxygalactonojirimycin) is a pharmacological chaperone that selectively binds and stabilizes α‐Gal A, increasing total cellular levels and activity for some mutant forms (defined as “responsive”). In this study, we developed a cell‐based assay in cultured HEK‐293 cells to identify mutant forms of α‐Gal A that are responsive to AT1001. Concentration‐dependent increases in α‐Gal A activity in response to AT1001 were shown for 49 (60%) of 81 mutant forms. The responses of α‐Gal A mutant forms were generally consistent with the responses observed in male Fabry patient‐derived lymphoblasts. Importantly, the HEK‐293 cell responses of 19 α‐Gal A mutant forms to a clinically achievable concentration of AT1001 (10 µM) were generally consistent with observed increases in α‐Gal A activity in peripheral blood mononuclear cells from male Fabry patients orally administered AT1001 during Phase 2 clinical studies. This indicates that the cell‐based responses can identify mutant forms of α‐Gal A that are likely to respond to AT1001 in vivo. Thus, the HEK‐293 cell‐based assay may be a useful aid in the identification of Fabry patients with AT1001‐responsive mutant forms. Hum Mutat 32:1–13, 2011.


Molecular Genetics and Metabolism | 2013

A Phase 2 study of migalastat hydrochloride in females with Fabry disease: Selection of population, safety and pharmacodynamic effects

Roberto Giugliani; Stephen Waldek; Dominique P. Germain; Kathy Nicholls; Daniel G. Bichet; J.K. Simosky; Alexander Bragat; Jeffrey P. Castelli; Elfrida R. Benjamin; Pol Boudes

BACKGROUND Fabry disease (FD) is a genetic disorder resulting from deficiency of the lysosomal enzyme α-galactosidase A (α-Gal A) which leads to globotriaosylceramide (GL-3) accumulation in multiple tissues. We report on the safety and pharmacodynamics of migalastat hydrochloride, an investigational pharmacological chaperone given orally every other day (QOD) to females with FD. METHODS This was an open-label, uncontrolled, Phase 2 study of 12 weeks with extension to 48 weeks in nine females with FD. Doses of 50mg, 150 mg and 250 mg were given QOD. At multiple time points, α-Gal A activity and GL-3 levels were quantified in blood cells, kidney and skin. GL-3 levels were also evaluated through skin and renal histology. Each individual GLA mutation was retrospectively categorized as being amenable or not to migalastat HCl based on an in vitro α-Gal A transfection assay developed in human embryonic kidney (HEK)-293 cells. RESULTS Migalastat HCl was generally well tolerated. Patients with amenable mutations seem to demonstrate greater pharmacodynamic response to migalastat HCl compared to patients with non-amenable mutations. The greatest declines in urine GL-3 were observed in the three patients with amenable GLA mutations that were treated with 150 or 250 mg migalastat HCl QOD. Additionally, these three patients all demonstrated decreases in GL-3 inclusions in kidney peri-tubular capillaries. CONCLUSIONS Migalastat HCl is a candidate oral pharmacological chaperone that provides a potential novel genotype-specific treatment for FD. Treatment resulted in GL-3 substrate decrease in female patients with amenable GLA mutations. Phase 3 studies are ongoing.


Journal of Medical Genetics | 2017

Oral pharmacological chaperone migalastat compared with enzyme replacement therapy in Fabry disease: 18-month results from the randomised phase III ATTRACT study

Derralynn Hughes; Kathleen Nicholls; Suma P. Shankar; Gere Sunder-Plassmann; David M. Koeller; Khan Nedd; Gerard Vockley; Takashi Hamazaki; Robin H. Lachmann; Toya Ohashi; Iacopo Olivotto; Norio Sakai; Patrick Deegan; David Dimmock; François Eyskens; Dominique P. Germain; Ozlem Goker-Alpan; Eric Hachulla; Ana Jovanovic; Charles Marques Lourenço; Ichiei Narita; Mark Thomas; William R. Wilcox; Daniel G. Bichet; Raphael Schiffmann; Elizabeth Ludington; Christopher Viereck; John Kirk; Julie Yu; Franklin K. Johnson

Background Fabry disease is an X-linked lysosomal storage disorder caused by GLA mutations, resulting in α-galactosidase (α-Gal) deficiency and accumulation of lysosomal substrates. Migalastat, an oral pharmacological chaperone being developed as an alternative to intravenous enzyme replacement therapy (ERT), stabilises specific mutant (amenable) forms of α-Gal to facilitate normal lysosomal trafficking. Methods The main objective of the 18-month, randomised, active-controlled ATTRACT study was to assess the effects of migalastat on renal function in patients with Fabry disease previously treated with ERT. Effects on heart, disease substrate, patient-reported outcomes (PROs) and safety were also assessed. Results Fifty-seven adults (56% female) receiving ERT (88% had multiorgan disease) were randomised (1.5:1), based on a preliminary cell-based assay of responsiveness to migalastat, to receive 18 months open-label migalastat or remain on ERT. Four patients had non-amenable mutant forms of α-Gal based on the validated cell-based assay conducted after treatment initiation and were excluded from primary efficacy analyses only. Migalastat and ERT had similar effects on renal function. Left ventricular mass index decreased significantly with migalastat treatment (−6.6 g/m2 (−11.0 to −2.2)); there was no significant change with ERT. Predefined renal, cardiac or cerebrovascular events occurred in 29% and 44% of patients in the migalastat and ERT groups, respectively. Plasma globotriaosylsphingosine remained low and stable following the switch from ERT to migalastat. PROs were comparable between groups. Migalastat was generally safe and well tolerated. Conclusions Migalastat offers promise as a first-in-class oral monotherapy alternative treatment to intravenous ERT for patients with Fabry disease and amenable mutations. Trial registration number: NCT00925301; Pre-results.


PLOS ONE | 2015

Oral Migalastat HCl Leads to Greater Systemic Exposure and Tissue Levels of Active α-Galactosidase A in Fabry Patients when Co-Administered with Infused Agalsidase

David G. Warnock; Daniel G. Bichet; Myrl Holida; Ozlem Goker-Alpan; Kathy Nicholls; Mark Thomas; François Eyskens; Suma P. Shankar; Mathews Adera; Sheela Sitaraman; Richie Khanna; John J. Flanagan; Brandon Wustman; Jay Barth; Carrolee Barlow; Kenneth J. Valenzano; David J. Lockhart; Pol Boudes; Franklin K. Johnson

Migalastat HCl (AT1001, 1-Deoxygalactonojirimycin) is an investigational pharmacological chaperone for the treatment of α-galactosidase A (α-Gal A) deficiency, which leads to Fabry disease, an X-linked, lysosomal storage disorder. The currently approved, biologics-based therapy for Fabry disease is enzyme replacement therapy (ERT) with either agalsidase alfa (Replagal) or agalsidase beta (Fabrazyme). Based on preclinical data, migalastat HCl in combination with agalsidase is expected to result in the pharmacokinetic (PK) enhancement of agalsidase in plasma by increasing the systemic exposure of active agalsidase, thereby leading to increased cellular levels in disease-relevant tissues. This Phase 2a study design consisted of an open-label, fixed-treatment sequence that evaluated the effects of single oral doses of 150 mg or 450 mg migalastat HCl on the PK and tissue levels of intravenously infused agalsidase (0.2, 0.5, or 1.0 mg/kg) in male Fabry patients. As expected, intravenous administration of agalsidase alone resulted in increased α-Gal A activity in plasma, skin, and peripheral blood mononuclear cells (PBMCs) compared to baseline. Following co-administration of migalastat HCl and agalsidase, α-Gal A activity in plasma was further significantly increased 1.2- to 5.1-fold compared to agalsidase administration alone, in 22 of 23 patients (95.6%). Importantly, similar increases in skin and PBMC α-Gal A activity were seen following co-administration of migalastat HCl and agalsidase. The effects were not related to the administered migalastat HCl dose, as the 150 mg dose of migalastat HCl increased α-Gal A activity to the same extent as the 450 mg dose. Conversely, agalsidase had no effect on the plasma PK of migalastat. No migalastat HCl-related adverse events or drug-related tolerability issues were identified. Trial Registration ClinicalTrials.gov NCT01196871


PLOS ONE | 2013

Migalastat HCl Reduces Globotriaosylsphingosine (Lyso-Gb3) in Fabry Transgenic Mice and in the Plasma of Fabry Patients

Brandy Young-Gqamana; Nastry Brignol; Hui-Hwa Chang; Richie Khanna; Rebecca Soska; Maria Fuller; Sheela Sitaraman; Dominique P. Germain; Roberto Giugliani; Derralynn Hughes; Atul Mehta; Kathy Nicholls; Pol Boudes; David J. Lockhart; Kenneth J. Valenzano; Elfrida R. Benjamin

Fabry disease (FD) results from mutations in the gene (GLA) that encodes the lysosomal enzyme α-galactosidase A (α-Gal A), and involves pathological accumulation of globotriaosylceramide (GL-3) and globotriaosylsphingosine (lyso-Gb3). Migalastat hydrochloride (GR181413A) is a pharmacological chaperone that selectively binds, stabilizes, and increases cellular levels of α-Gal A. Oral administration of migalastat HCl reduces tissue GL-3 in Fabry transgenic mice, and in urine and kidneys of some FD patients. A liquid chromatography-tandem mass spectrometry method was developed to measure lyso-Gb3 in mouse tissues and human plasma. Oral administration of migalastat HCl to transgenic mice reduced elevated lyso-Gb3 levels up to 64%, 59%, and 81% in kidney, heart, and skin, respectively, generally equal to or greater than observed for GL-3. Furthermore, baseline plasma lyso-Gb3 levels were markedly elevated in six male FD patients enrolled in Phase 2 studies. Oral administration of migalastat HCl (150 mg QOD) reduced urine GL-3 and plasma lyso-Gb3 in three subjects (range: 15% to 46% within 48 weeks of treatment). In contrast, three showed no reductions in either substrate. These results suggest that measurement of tissue and/or plasma lyso-Gb3 is feasible and may be warranted in future studies of migalastat HCl or other new potential therapies for FD.


Clinical pharmacology in drug development | 2013

Pharmacokinetics and Safety of Migalastat HCl and Effects on Agalsidase Activity in Healthy Volunteers

Franklin K. Johnson; Paul N. Mudd; Alexander Bragat; Mathews Adera; Pol Boudes

Migalastat HCl is an investigational, oral treatment for Fabry disease, an X‐linked lysosomal storage disorder. Four Phase 1 studies were conducted to determine the pharmacokinetics, pharmacodynamics, safety, and tolerability of migalastat. Healthy volunteers (N = 124), 18–55 years old, received migalastat HCl single (25 mg–2000 mg) or twice‐daily doses (50 mg, 150 mg) for 7 days in a double‐blind, placebo‐controlled fashion. Migalastat pharmacokinetics were dose‐proportional (AUC∞ range: 1129–72 838 ng h/mL, Cmax range: 200.5–13 844 ng/mL, t1/2 3–4 hours). Steady state was achieved by Day 7. Up to 67% of the dose was excreted as unchanged drug in urine. Increased α‐Gal A activity was dose related. No abnormal cardiac effects, including prolonged QTc intervals, were observed. The pharmacokinetics of migalastat were well characterized in these Phase 1 studies conducted healthy volunteers. The 150 mg dose of migalastat HCl administered BID for 7 days was generally safe and well tolerated. A TQT study demonstrated lack of a positive signal at therapeutic and supra‐therapeutic doses. Increases in α‐Gal A enzyme activity for the 150 mg dose observed in healthy subjects suggested a successful proof of mechanism for further investigations.


Archives of Pathology & Laboratory Medicine | 2012

Novel Quantitative Method to Evaluate Globotriaosylceramide Inclusions in Renal Peritubular Capillaries by Virtual Microscopy in Patients With Fabry Disease

Laura Barisoni; J. Charles Jennette; Robert B. Colvin; Sheela Sitaraman; Alexander Bragat; Jeff Castelli; Dan Walker; Pol Boudes

CONTEXT Assessing the amount of globotriaosylceramide inclusions in renal peritubular capillaries by a semiquantitative approach is a standard and useful measure of therapeutic efficacy in Fabry disease, achievable by light microscopy analysis. OBJECTIVE To describe a novel virtual microscopy quantitative method to measure globotriaosylceramide inclusions (Barisoni Lipid Inclusion Scoring System [BLISS]) in renal biopsies from patients with Fabry disease. DESIGN Plastic embedded 1-µm-thick sections from kidney biopsies from 17 patients enrolled in a Fabry disease clinical trial were evaluated using a standard semiquantitative methodology and BLISS to compare sensitivity. We also tested intrareader and interreader variability of BLISS and compared results from conventional light microscopy analysis with a virtual microscopy-based methodology. Peritubular capillaries were first annotated on digital images of whole slides by 1 pathologist and then scored for globotriaosylceramide inclusions by 2 additional pathologists. RESULTS We demonstrated that (1) quantitative analysis by BLISS results in detection of small amount of globotriaosylceramide inclusions even when by semiquantitative analysis the score is 0, (2) application of BLISS combined with conventional light microscopy results in low intrareader and interreader variability, and (3) BLISS combined with virtual microscopy results in significant reduction of intrareader and interreader variability compared with BLISS-light microscopy. CONCLUSIONS BLISS is a simpler and more sensitive scoring system compared to the semiquantitative approach. The virtual microscopy-based methodology increases accuracy and reproducibility; moreover, it provides a permanent record of retrievable data with full transparency in clinical trials.


Molecular Therapy | 2017

Duvoglustat HCl Increases Systemic and Tissue Exposure of Active Acid α-Glucosidase in Pompe Patients Co-administered with Alglucosidase α

Priya S. Kishnani; Mark A. Tarnopolsky; Mark Roberts; K. Sivakumar; Majed Dasouki; Mazen M. Dimachkie; Erika L. Finanger; Ozlem Goker-Alpan; Karl Guter; Tahseen Mozaffar; Muhammad Ali Pervaiz; P. Laforêt; Todd Levine; Matthews Adera; Richard Lazauskas; Sheela Sitaraman; Richie Khanna; Elfrida R. Benjamin; Jessie Feng; John J. Flanagan; Jay Barth; Carrolee Barlow; David J. Lockhart; Kenneth J. Valenzano; Pol Boudes; Franklin K. Johnson; Barry J. Byrne

Duvoglustat HCl (AT2220, 1-deoxynojirimycin) is an investigational pharmacological chaperone for the treatment of acid α-glucosidase (GAA) deficiency, which leads to the lysosomal storage disorder Pompe disease, which is characterized by progressive accumulation of lysosomal glycogen primarily in heart and skeletal muscles. The current standard of care is enzyme replacement therapy with recombinant human GAA (alglucosidase alfa [AA], Genzyme). Based on preclinical data, oral co-administration of duvoglustat HCl with AA increases exposure of active levels in plasma and skeletal muscles, leading to greater substrate reduction in muscle. This phase 2a study consisted of an open-label, fixed-treatment sequence that evaluated the effect of single oral doses of 50 mg, 100 mg, 250 mg, or 600 mg duvoglustat HCl on the pharmacokinetics and tissue levels of intravenously infused AA (20 mg/kg) in Pompe patients. AA alone resulted in increases in total GAA activity and protein in plasma compared to baseline. Following co-administration with duvoglustat HCl, total GAA activity and protein in plasma were further increased 1.2- to 2.8-fold compared to AA alone in all 25 Pompe patients; importantly, muscle GAA activity was increased for all co-administration treatments from day 3 biopsy specimens. No duvoglustat-related adverse events or drug-related tolerability issues were identified.


Clinical pharmacology in drug development | 2015

An open‐label study to determine the pharmacokinetics and safety of migalastat HCl in subjects with impaired renal function and healthy subjects with normal renal function

Franklin K. Johnson; Paul N. Mudd; Tara L. Dimino; Jennie Vosk; Sheela Sitaraman; Pol Boudes; Carrolee Barlow

Renal function may progressively decline in patients with Fabry disease. This study assessed pharmacokinetics, safety, and tolerability of a single oral dose of migalastat HCl 150 mg in subjects with normal or mildly, moderately, or severely impaired renal function.

Collaboration


Dive into the Pol Boudes's collaboration.

Top Co-Authors

Avatar

Kathy Nicholls

Royal Melbourne Hospital

View shared research outputs
Top Co-Authors

Avatar

Ozlem Goker-Alpan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roberto Giugliani

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge