Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
N Engl J Med ; 390(18): 1663-1676, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38657265

ABSTRACT

BACKGROUND: Exagamglogene autotemcel (exa-cel) is a nonviral cell therapy designed to reactivate fetal hemoglobin synthesis through ex vivo clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene editing of the erythroid-specific enhancer region of BCL11A in autologous CD34+ hematopoietic stem and progenitor cells (HSPCs). METHODS: We conducted an open-label, single-group, phase 3 study of exa-cel in patients 12 to 35 years of age with transfusion-dependent ß-thalassemia and a ß0/ß0, ß0/ß0-like, or non-ß0/ß0-like genotype. CD34+ HSPCs were edited by means of CRISPR-Cas9 with a guide mRNA. Before the exa-cel infusion, patients underwent myeloablative conditioning with pharmacokinetically dose-adjusted busulfan. The primary end point was transfusion independence, defined as a weighted average hemoglobin level of 9 g per deciliter or higher without red-cell transfusion for at least 12 consecutive months. Total and fetal hemoglobin concentrations and safety were also assessed. RESULTS: A total of 52 patients with transfusion-dependent ß-thalassemia received exa-cel and were included in this prespecified interim analysis; the median follow-up was 20.4 months (range, 2.1 to 48.1). Neutrophils and platelets engrafted in each patient. Among the 35 patients with sufficient follow-up data for evaluation, transfusion independence occurred in 32 (91%; 95% confidence interval, 77 to 98; P<0.001 against the null hypothesis of a 50% response). During transfusion independence, the mean total hemoglobin level was 13.1 g per deciliter and the mean fetal hemoglobin level was 11.9 g per deciliter, and fetal hemoglobin had a pancellular distribution (≥94% of red cells). The safety profile of exa-cel was generally consistent with that of myeloablative busulfan conditioning and autologous HSPC transplantation. No deaths or cancers occurred. CONCLUSIONS: Treatment with exa-cel, preceded by myeloablation, resulted in transfusion independence in 91% of patients with transfusion-dependent ß-thalassemia. (Supported by Vertex Pharmaceuticals and CRISPR Therapeutics; CLIMB THAL-111 ClinicalTrials.gov number, NCT03655678.).


Subject(s)
Fetal Hemoglobin , Gene Editing , Hematopoietic Stem Cell Transplantation , beta-Thalassemia , Adolescent , Adult , Child , Female , Humans , Male , Young Adult , Antigens, CD34 , beta-Thalassemia/therapy , beta-Thalassemia/genetics , Blood Transfusion , Busulfan/therapeutic use , CRISPR-Cas Systems , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Gene Editing/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells , Repressor Proteins/genetics , Transplantation Conditioning , Transplantation, Autologous , Myeloablative Agonists/therapeutic use , North America , Europe
2.
Drug Saf ; 45(2): 127-136, 2022 02.
Article in English | MEDLINE | ID: mdl-35020177

ABSTRACT

INTRODUCTION: Approved in 1994 and assigned the International Nonproprietary Name (INN) imiglucerase by the World Health Organization, Cerezyme® (Sanofi Genzyme) is an enzyme replacement therapy used to treat Gaucher disease in > 90 countries. At least two therapies approved outside the USA and the European Union, Abcertin® and Asbroder®, have adopted the identical INN imiglucerase. Both drugs were approved via regulatory pathways not aligned with World Health Organization Similar Biotherapeutic Product guidelines. OBJECTIVE: We analyzed whether the use of the identical INN "imiglucerase" for these drugs impacts adverse event (AE) reporting in the Sanofi Global Safety Database. METHODS: First, we reviewed all imiglucerase individual case safety reports (referred to as cases) including AE data reported between January 2012 and March 2018 that contained Abcertin or Asbroder in the narrative. In a second analysis, we examined cases from Mexico reported between May 2013 and March 2018 to assess changes in imiglucerase reporting following the 2015 approval of Asbroder in Mexico. RESULTS: Fifty-six cases mentioning Asbroder and none mentioning Abcertin were retrieved in the first analysis. Upon close review, the AEs of 45 cases (80.4%) were attributed to Asbroder, one (1.8%) to Cerezyme; the specific drug attribution for the AEs of ten cases (17.9%) could not be determined. In the second analysis, a substantial increase in cases and AEs was observed in the period after Asbroder approval (73 cases with 150 AEs pre-approval vs 132 cases with 333 AEs post-approval). Twenty-three of 132 (17.4%) post-approval cases reported discontinuation of treatment (19 related to Asbroder AEs, and four related to Cerezyme AEs). Infusion-associated reactions occurred in 25/132 cases (17 Asbroder related, six Cerezyme related, two indeterminate). CONCLUSIONS: This analysis demonstrates two potential consequences of identical INN use between Cerezyme and Asbroder: (1) an aggregate safety profile for Cerezyme that includes other products using the identical INN leading to inaccurate pharmacovigilance data and (2) healthcare providers switching, substituting, or potentially assuming interchangeability between the products. Identical INN use without the brand name differentiator may compromise pharmacovigilance data, potentially masking differences in safety profiles between products.


The objective of this study was to assess the consequences of multiple drugs using the identical International Nonproprietary Name (INN) "imiglucerase" on adverse event reporting in the Sanofi Genzyme Global Safety Database. The World Health Organization established the INN system to identify drugs that are made of the same pharmaceutical substance and recommends that different products have distinct INN names. The INN imiglucerase was assigned in 1994 to Cerezyme® (Sanofi Genzyme), an orphan drug for the treatment of a rare disease known as Gaucher disease. In 2015, Asbroder® (ISU Abxis) was approved for Gaucher disease in Mexico and has adopted the INN imiglucerase. It was not approved as a biosimilar to Cerezyme. Most importantly, in a significant proportion of the adverse event cases reported, patients received a combination therapy of Asbroder and Cerezyme or Asbroder and "imiglucerase", suggesting that the shared INN may have led to misconceived interchangeability of these products. Such confusion among healthcare providers poses a potentially serious risk to patient safety and health. These results are especially worrisome because they relate to products sharing an INN that were not approved as biosimilars. The findings from this study are also consistent with the view that Cerezyme and Asbroder may have different safety profiles. The implications of drug products having the same INN are discussed in the article as well as recommended solutions. To our knowledge, this is one of the first reports on real-world safety experience with biologics sharing the same INN name.


Subject(s)
Gaucher Disease , Databases, Factual , European Union , Gaucher Disease/drug therapy , Glucosylceramidase , Humans , Pharmacovigilance
3.
Mol Genet Metab ; 131(1-2): 211-218, 2020.
Article in English | MEDLINE | ID: mdl-33012655

ABSTRACT

Eliglustat is a first-line oral treatment for adults with Gaucher disease type 1 who have cytochrome P450 (CYP) 2D6 extensive, intermediate, or poor metabolizer phenotypes. Per International Conference on Harmonisation (ICH) E14 guidance, a Phase 1 thorough electrocardiographic (ECG) study was done during drug development to assess eliglustat's effects on cardiac repolarization by measuring ECG intervals in healthy adult subjects. Using data from the thorough ECG study, we performed pharmacokinetic/pharmacodynamic-ECG modeling to establish the relationship between eliglustat concentrations and their effects on ECG intervals. We then used that concentration-response relationship to predict the effects of eliglustat on each ECG interval for each CYP2D6 metabolizer phenotype (the main determinant of eliglustat exposure) and in different drug-drug interaction scenarios. These predictions, together with other exposure-related factors, contributed to the CYP2D6 phenotype-based dosing recommendations for eliglustat, including dose adjustments and contraindications when co-administered with drugs metabolized by the CYP2D6 and CYP3A pathways.


Subject(s)
Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP3A/genetics , Gaucher Disease/drug therapy , Pyrrolidines/administration & dosage , Administration, Oral , Adult , Dose-Response Relationship, Drug , Drug Interactions/genetics , Electrocardiography , Female , Gaucher Disease/genetics , Gaucher Disease/pathology , Humans , Inactivation, Metabolic/genetics , Liver/drug effects , Male , Pyrrolidines/pharmacokinetics
4.
Mol Genet Metab ; 129(4): 278-285, 2020 04.
Article in English | MEDLINE | ID: mdl-32029355

ABSTRACT

Eliglustat is an oral substrate reduction therapy indicated for patients with Gaucher disease type 1. Based on in vitro data, clinical trials were conducted to assess the potential for drug-drug interactions between eliglustat and digoxin (P-glycoprotein substrate), metoprolol (sensitive CYP2D6 substrate), a combined oral contraceptive (CYP3A substrate), and acid-reducing agents. Healthy subjects were enrolled in four Phase 1 clinical studies to evaluate the effect of eliglustat on the pharmacokinetics, safety, and tolerability of digoxin (N = 28), metoprolol (N = 14), and a combined oral contraceptive (N = 30) and the effect of acid-reducing agents on eliglustat pharmacokinetics, safety, and tolerability (N = 24). Coadministration resulted in increased exposure to digoxin (1.49-fold) and metoprolol (2-fold) with eliglustat, negligible effects on oral contraceptive pharmacokinetics with eliglustat, and a negligible effect of acid-reducing agents on eliglustat pharmacokinetics. Across all studies, eliglustat was well-tolerated. One serious adverse event (spontaneous abortion) and one discontinuation due to an adverse event (urinary tract infection) were reported, both during the acid-reducing agents study. When eliglustat is coadministered with medications that are P-glycoprotein or CYP2D6 substrates, lower doses of these concomitant medications may be required. Eliglustat may be coadministered with oral contraceptives and acid-reducing agents without dose modifications for either drug.


Subject(s)
Contraceptives, Oral/pharmacokinetics , Digoxin/pharmacokinetics , Metoprolol/pharmacokinetics , Proton Pump Inhibitors/administration & dosage , Pyrrolidines/administration & dosage , Adolescent , Adult , Contraceptives, Oral/administration & dosage , Contraceptives, Oral/adverse effects , Cross-Over Studies , Digoxin/administration & dosage , Digoxin/adverse effects , Drug Interactions , Female , Humans , Male , Metoprolol/administration & dosage , Metoprolol/adverse effects , Middle Aged , Proton Pump Inhibitors/adverse effects , Young Adult
5.
Mol Genet Metab Rep ; 22: 100552, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31993325

ABSTRACT

Eliglustat is an oral glucosylceramide synthase inhibitor indicated for the long-term treatment of adults with Gaucher disease type 1 and CYP2D6 extensive, intermediate, or poor metabolizer phenotypes. Eliglustat is metabolized primarily by CYP2D6 and to a lesser extent by CYP3A4 and is a substrate of P-glycoprotein (P-gp). Three studies evaluated the effects of paroxetine (strong CYP2D6 inhibitor), ketoconazole (strong CYP3A4 and P-gp inhibitor), and rifampin (strong CYP3A4/P-gp inducer; OATP inhibitor) on the pharmacokinetics of orally administered eliglustat in healthy adults. An 8.9-fold increase in eliglustat exposure following co-administration of multiple-dose eliglustat and paroxetine is attributed to inhibition of CYP2D6-mediated metabolism of eliglustat by paroxetine. A 4.3-fold increase in eliglustat exposure following co-administration of multiple-dose eliglustat and ketoconazole is attributed to inhibition of CYP3A4-mediated metabolism and/or P-gp-mediated transport of eliglustat by ketoconazole. Co-administration of eliglustat with oral doses of rifampin reduced eliglustat exposure by >85% due to induction of CYP3A4/P-gp by rifampin, while a single intravenous dose of rifampin had no effect on eliglustat, confirming that eliglustat is not an OATP substrate. Depending on CYP2D6 metabolizer phenotype, co-administration of eliglustat with CYP2D6 and/or CYP3A inhibitors or CYP3A inducers may alter eliglustat exposure, warrant dosage adjustment or use with caution, or be contraindicated.

6.
Lancet ; 385(9985): 2355-62, 2015 Jun 13.
Article in English | MEDLINE | ID: mdl-25819691

ABSTRACT

BACKGROUND: The mainstay of treatment for Gaucher's disease type 1 is alternate-week infusion of enzyme replacement therapy (ERT). We investigated whether patients stable on such treatment would remain so after switching to oral eliglustat, a selective inhibitor of glucosylceramide synthase. METHODS: In this phase 3, randomised, multinational, open-label, non-inferiority trial, we enrolled adults (aged ≥18 years) who had received ERT for 3 years or more for Gaucher's disease. Patients were randomly allocated 2:1 at 39 clinics (stratified by ERT dose; block sizes of four; computer-generated centrally) to receive either oral eliglustat or imiglucerase infusions for 12 months. Participants and investigators were aware of treatment assignment, but the central reader who assessed organ volumes was masked. The composite primary efficacy endpoint was percentage of patients whose haematological variables and organ volumes remained stable for 12 months (ie, haemoglobin decrease not more than 15 g/L, platelet count decrease not more than 25%, spleen volume increase not more than 25%, and liver volume increase not more than 20%, in multiples of normal from baseline). The non-inferiority margin was 25% for eliglustat relative to imiglucerase, assessed in all patients who completed 12 months of treatment. This trial is registered with ClinicalTrials.gov, number NCT00943111, and EudraCT, number 2008-005223-28. FINDINGS: Between Sept 15, 2009, and Nov 9, 2011, we randomly allocated 106 (66%) patients to eliglustat and 54 (34%) to imiglucerase. In the per-protocol population, 84 (85%) of 99 patients who completed eliglustat treatment and 44 (94%) of 47 patients who completed imiglucerase treatment met the composite primary endpoint (between-group difference -8·8%; 95% CI -17·6 to 4·2). The lower bound of the 95% CI of -17·6% was within the prespecified threshold for non-inferiority. Dropouts occurred due to palpitations (one patient on eliglustat), myocardial infarction (one patient on eliglustat), and psychotic disorder (one patient on imiglucerase). No deaths occurred. 97 (92%) of 106 patients in the eliglustat group had treatment-emergent adverse events, as did 42 (79%) of 53 in the imiglucerase group (mostly mild or moderate in severity). INTERPRETATION: Oral eliglustat maintained haematological and organ volume stability in adults with Gaucher's disease type 1 already controlled by intravenous ERT and could be a useful therapeutic option. FUNDING: Genzyme, a Sanofi company.


Subject(s)
Enzyme Inhibitors/therapeutic use , Enzyme Replacement Therapy , Gaucher Disease/drug therapy , Glucosylceramidase/therapeutic use , Pyrrolidines/therapeutic use , Administration, Oral , Adult , Female , Gaucher Disease/blood , Hemoglobins/analysis , Humans , Infusions, Intravenous , Liver/pathology , Magnetic Resonance Imaging , Male , Organ Size , Platelet Count , Spleen/pathology
7.
JAMA ; 313(7): 695-706, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25688781

ABSTRACT

IMPORTANCE: Gaucher disease type 1 is characterized by hepatosplenomegaly, anemia, thrombocytopenia, and skeletal disease. A safe, effective oral therapy is needed. OBJECTIVE: To determine whether eliglustat, a novel oral substrate reduction therapy, safely reverses clinical manifestations in untreated adults with Gaucher disease type 1. DESIGN, SETTING, AND PARTICIPANTS: Phase 3, randomized, double-blind, placebo-controlled trial conducted at 18 sites in 12 countries from November 2009 to July 2012 among eligible patients with splenomegaly plus thrombocytopenia and/or anemia. Of 72 patients screened, 40 were enrolled. INTERVENTIONS: Patients were stratified by spleen volume and randomized 1:1 to receive eliglustat (50 or 100 mg twice daily; n = 20) or placebo (n = 20) for 9 months. MAIN OUTCOMES AND MEASURES: The primary efficacy end point was percentage change in spleen volume in multiples of normal from baseline to 9 months; secondary efficacy end points were change in hemoglobin level and percentage changes in liver volume and platelet count. RESULTS: All patients had baseline splenomegaly and thrombocytopenia (mostly moderate or severe), most had mild or moderate hepatomegaly, and 20% had mild anemia. Least-square mean spleen volume decreased by 27.77% (95% CI, -32.57% to -22.97%) in the eliglustat group (from 13.89 to 10.17 multiples of normal) vs an increase of 2.26% (95% CI, -2.54% to 7.06%) in the placebo group (from 12.50 to 12.84 multiples of normal) for an absolute treatment difference of -30.03% (95% CI, -36.82% to -23.24%; P < .001). For the secondary end points, the least-square mean absolute differences between groups all favored eliglustat, with a 1.22-g/dL increase in hemoglobin level (95% CI, 0.57-1.88 g/dL; P < .001), 6.64% decrease in liver volume (95% CI, -11.37% to -1.91%; P = .007), and 41.06% increase in platelet count (95% CI, 23.95%-58.17%; P < .001). No serious adverse events occurred. One patient in the eliglustat group withdrew (non-treatment related); 39 of the 40 patients transitioned to an open-label extension study. CONCLUSIONS AND RELEVANCE: Among previously untreated adults with Gaucher disease type 1, treatment with eliglustat compared with placebo for 9 months resulted in significant improvements in spleen volume, hemoglobin level, liver volume, and platelet count. The clinical significance of these findings is uncertain, and more definitive conclusions about clinical efficacy and utility will require comparison with the standard treatment of enzyme replacement therapy as well as longer-term follow-up. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00891202.


Subject(s)
Enzyme Inhibitors/therapeutic use , Gaucher Disease/drug therapy , Glucosyltransferases/antagonists & inhibitors , Pyrrolidines/therapeutic use , Splenomegaly/drug therapy , Administration, Oral , Adult , Double-Blind Method , Enzyme Inhibitors/pharmacology , Female , Gaucher Disease/complications , Humans , Male , Organ Size/drug effects , Pyrrolidines/pharmacology , Spleen/pathology , Splenomegaly/etiology , Young Adult
8.
Blood Cells Mol Dis ; 53(4): 274-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24835462

ABSTRACT

Eliglustat is an investigational, oral substrate reduction therapy for Gaucher disease type 1 (GD1). Nineteen treatment-naïve patients have now completed 4years of an open-label study (NCT00358150). Mean hemoglobin level and platelet count increased by 2.3±1.5g/dL (baseline: 11.3±1.5g/dL) and 95% (baseline: 68,700±21,200/mm(3)), respectively. Mean spleen and liver volumes (multiples of normal, MN) decreased by 63% (baseline: 17.3±9.5 MN) and 28% (baseline: 1.7±0.4 MN), respectively. Median chitotriosidase and CCL-18 each decreased by 82%; plasma glucosylceramide and GM3 normalized. Mean bone mineral density T-score for the lumbar spine increased by 0.8 (60%) (baseline: -1.6±1.1). Femur dark marrow, a reflection of Gaucher cell infiltration into bone marrow, was reduced or stable in 17/18 patients. There were no bone crises. Most adverse events were mild and unrelated to treatment. These results extend the safety and efficacy of eliglustat reported at 1 and 2 years to 4 years.


Subject(s)
Drugs, Investigational/therapeutic use , Enzyme Inhibitors/therapeutic use , Gaucher Disease/drug therapy , Pyrrolidines/therapeutic use , Adolescent , Adult , Bone Density , Bone Marrow/drug effects , Bone Marrow/pathology , Chemokines, CC/blood , Female , Follow-Up Studies , G(M3) Ganglioside/blood , Gaucher Disease/blood , Gaucher Disease/pathology , Glucosylceramides/blood , Hemoglobins/metabolism , Hexosaminidases/blood , Humans , Lumbar Vertebrae/drug effects , Male , Middle Aged , Platelet Count
9.
Clin Cancer Res ; 9(15): 5721-8, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14654557

ABSTRACT

Ovarian cancer is the most lethal gynecological malignancy and the fifth most common cause of cancer in women. It is characterized by diffuse peritoneal carcinomatosis and often by large volumes of i.p. ascites. Because vascular endothelial growth factor (VEGF), also known as vascular permeability factor, increases vascular permeability and stimulates endothelial cell growth, its role in ovarian cancer has been evaluated in a number of studies. However, questions remain regarding the ability of VEGF alone to cause ascites formation and the ability of VEGF blockade to inhibit the growth of disseminated cancer. We have used retroviral technology to create cell populations that overproduce VEGF and report that enforced expression of VEGF by ovarian carcinoma cells dramatically reduces the time to onset of ascites formation. In fact, even tumor-free peritoneal overexpression of VEGF, created by using adenoviral vectors, is sufficient to cause ascites to accumulate. We have found that systemic administration of the VEGF-Trap, a recently described high-affinity soluble decoy receptor for VEGF, prevents ascites accumulation and also inhibits the growth of disseminated cancer. Remarkably, much as is observed in s.c. tumor models, VEGF blockade results in dramatic remodeling of the blood vessels in disseminated ovarian carcinoma. The potent effects of the VEGF-Trap in reducing both ascites and tumor burden suggest that it will be of value in a regimen for treatment of women with ovarian cancer and ascites.


Subject(s)
Ascites/prevention & control , Neovascularization, Pathologic/prevention & control , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/pathology , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/drug therapy , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...