Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Mol Ther ; 2024 May 24.
Article En | MEDLINE | ID: mdl-38796703

Gene transfer therapies utilizing adeno-associated virus (AAV) vectors involve a complex drug design with multiple components that may impact immunogenicity. Valoctocogene roxaparvovec is an AAV serotype 5 (AAV5)-vectored gene therapy for treatment of hemophilia A that encodes a B-domain-deleted human factor VIII (FVIII) protein controlled by a hepatocyte-selective promoter. Following previous results from the first-in-human phase 1/2 clinical trial, we assessed AAV5 capsid- and transgene-derived FVIII-specific immune responses with 2 years of follow-up data from GENEr8-1, a phase 3, single-arm, open-label study in 134 adult men with severe hemophilia A. No FVIII inhibitors were detected following administration of valoctocogene roxaparvovec. Immune responses were predominantly directed toward the AAV5 capsid, with all participants developing durable anti-AAV5 antibodies. Cellular immune responses specific for the AAV5 capsid were detected in most participants by interferon-ᵧ enzyme-linked immunosorbent spot assay 2 weeks following dose administration and declined or reverted to negative over the first 52 weeks. These responses were weakly correlated with alanine aminotransferase elevations and showed no association with changes in FVIII activity. FVIII-specific cellular immune responses were less frequent and more sporadic compared with those specific for AAV5 and showed no association with safety or efficacy parameters.

2.
Sci Rep ; 11(1): 24278, 2021 12 20.
Article En | MEDLINE | ID: mdl-34930956

Evidence from genetic disorders of CNP signalling suggests that plasma concentrations of CNP are subject to feedback regulation. In subjects with Achondroplasia (Ach), CNP intracellular activity is suppressed and plasma concentrations are raised but the therapeutic impact of exogenous CNP agonists on endogenous CNP is unknown. In this exploratory dose finding and extension study of 28 Ach children receiving Vosoritide over a 5 year period of treatment, endogenous CNP production was assessed using measurements of plasma aminoterminal proCNP (NTproCNP) adjusted for age and sex and normalised as standard deviation score (SDS), and then related to skeletal growth. Before treatment NTproCNP SDS was raised. Within the first 3 months of accelerating growth, levels were significantly reduced. Across the 5 years of sustained growth, levels varied widely and were markedly increased in some subjects during adolescence. Plasma NTproCNP was suppressed at 4 h post-injection in proportion to the prevailing level of hormone resistance as reflected by SDS before injection. We conclude CNP remains subject to regulation during growth promoting doses of Vosoritide. Fall in CNP during accelerating growth is consistent with an indirect feedback whereas the fall at 4 h is likely to be a direct effect from removal of intra cellular CNP resistance.


Achondroplasia/drug therapy , Natriuretic Peptide, C-Type/analogs & derivatives , Natriuretic Peptide, C-Type/metabolism , Adolescent , Bone Development/drug effects , Bone and Bones/drug effects , Child , Child, Preschool , Feedback , Female , Growth Disorders/drug therapy , Humans , Male , Natriuretic Peptide, C-Type/pharmacology , Time Factors , Treatment Outcome
3.
Genet Med ; 23(12): 2443-2447, 2021 12.
Article En | MEDLINE | ID: mdl-34341520

PURPOSE: Achondroplasia is caused by pathogenic variants in the fibroblast growth factor receptor 3 gene that lead to impaired endochondral ossification. Vosoritide, an analog of C-type natriuretic peptide, stimulates endochondral bone growth and is in development for the treatment of achondroplasia. This phase 3 extension study was conducted to document the efficacy and safety of continuous, daily vosoritide treatment in children with achondroplasia, and the two-year results are reported. METHODS: After completing at least six months of a baseline observational growth study, and 52 weeks in a double-blind, placebo-controlled study, participants were eligible to continue treatment in an open-label extension study, where all participants received vosoritide at a dose of 15.0 µg/kg/day. RESULTS: In children randomized to vosoritide, annualized growth velocity increased from 4.26 cm/year at baseline to 5.39 cm/year at 52 weeks and 5.52 cm/year at week 104. In children who crossed over from placebo to vosoritide in the extension study, annualized growth velocity increased from 3.81 cm/year at week 52 to 5.43 cm/year at week 104. No new adverse effects of vosoritide were detected. CONCLUSION: Vosoritide treatment has safe and persistent growth-promoting effects in children with achondroplasia treated daily for two years.


Achondroplasia , Natriuretic Peptide, C-Type , Achondroplasia/drug therapy , Achondroplasia/genetics , Child , Double-Blind Method , Humans , Natriuretic Peptide, C-Type/analogs & derivatives , Natriuretic Peptide, C-Type/therapeutic use , Treatment Outcome
4.
Thromb Haemost ; 117(6): 1093-1100, 2017 06 02.
Article En | MEDLINE | ID: mdl-28382371

Dyspnoea may be induced by some reversibly-binding P2Y12 inhibitors, including cangrelor and ticagrelor. Dyspnoea was not associated with any compromise to the efficacy of ticagrelor in the PLATO study. The CHAMPION PHOENIX study (NCT01156571) compared initial treatment with cangrelor versus initial treatment with clopidogrel in patients undergoing PCI. We investigated the incidence, characteristics, and associated clinical outcomes in patients with dyspnoea in CHAMPION PHOENIX. Adverse events (AEs) of dyspnoea to 48 hours were recorded in patients randomised to cangrelor or clopidogrel in CHAMPION PHOENIX. The composite primary endpoint of death, myocardial infarction, ischaemia-driven revascularisation, or stent thrombosis as well its individual components were assessed in patients who did or did not report dyspnoea. A total of 68 (1.2 %) cangrelor-treated patients and 18 (0.3 %) clopidogrel-treated patients reported dyspnoea (p<0.001). Most dyspnoea events in cangrelor-treated patients were considered mild (71 %) or moderate (28 %) and only one event was considered severe and led to discontinuation of cangrelor. The dyspnoea events in the clopidogrel-treated patients were mild (78 %) or moderate (22 %). Characteristics of dyspnoea were consistent with those seen in the CHAMPION programme as a whole. In the modified intention-to-treat population, rates of the composite primary outcome and its individual components were not affected by the presence of dyspnoea in cangrelor-treated patients. Cangrelor-related dyspnoea is transient, usually mild or moderate, and unlikely to lead to discontinuation of therapy. The occurrence of dyspnoea does not seem to be associated with any reduction in the efficacy of cangrelor compared with clopidogrel as initial therapy in PCI patients.


Adenosine Monophosphate/analogs & derivatives , Coronary Occlusion/epidemiology , Drug-Related Side Effects and Adverse Reactions/epidemiology , Dyspnea/epidemiology , Ticlopidine/analogs & derivatives , Adenosine Monophosphate/therapeutic use , Aged , Clopidogrel , Coronary Occlusion/drug therapy , Coronary Occlusion/mortality , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions/mortality , Dyspnea/mortality , Female , Humans , Intention to Treat Analysis , Male , Middle Aged , Percutaneous Coronary Intervention , Receptors, Purinergic P2Y12/metabolism , Survival Analysis , Ticlopidine/therapeutic use , Treatment Outcome , Withholding Treatment
5.
J Thorac Cardiovasc Surg ; 131(1): 21-7, 2006 Jan.
Article En | MEDLINE | ID: mdl-16399290

OBJECTIVE: Thrombin is generated in significant quantities during cardiopulmonary bypass and mediates adverse events, such as platelet aggregation and proinflammatory responses, through activation of the high-affinity thrombin receptor protease-activated receptor 1, which is expressed on platelets and endothelium. Thus antagonism of protease-activated receptor 1 might have broad therapeutic significance. Aprotinin, used clinically to reduce transfusion requirements and the inflammatory response to bypass, has been shown to inhibit protease-activated receptor 1 on platelets in vitro and in vivo. Here we have examined whether aprotinin inhibits endothelial protease-activated receptor 1 activation and resulting proinflammatory responses induced by thrombin. METHODS: Protease-activated receptor 1 expression and function were examined in cultured human umbilical vein endothelial cells after treatment with alpha-thrombin at 0.02 to 0.15 U/mL in the presence or absence of aprotinin (200-1600 kallikrein inhibitory units/mL). Protease-activated receptor 1 activation was assessed by using an antibody, SPAN-12, which detects only the unactivated receptor, and thrombin-mediated calcium fluxes. Other thrombin-dependent inflammatory pathways investigated were phosphorylation of the p42/44 mitogen-activated protein kinase, upregulation of the early growth response 1 transcription factor, and production of the proinflammatory cytokine interleukin 6. RESULTS: Pretreatment of cultured endothelial cells with aprotinin significantly spared protease-activated receptor 1 receptor cleavage (P < .0001) and abrogated calcium fluxes caused by thrombin. Aprotinin inhibited intracellular signaling through p42/44 mitogen-activated protein kinase (P < .05) and early growth response 1 transcription factor (P < .05), as well as interleukin 6 secretion caused by thrombin (P < .005). CONCLUSIONS: This study demonstrates that endothelial cell activation by thrombin and downstream inflammatory responses can be inhibited by aprotinin in vitro through blockade of protease-activated receptor 1. Our results provide a new molecular basis to help explain the anti-inflammatory properties of aprotinin reported clinically.


Aprotinin/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Receptor, PAR-1/antagonists & inhibitors , Thrombin/physiology , Aprotinin/therapeutic use , Cells, Cultured , Humans , Inflammation/prevention & control
6.
Ann Thorac Surg ; 81(2): 619-24, 2006 Feb.
Article En | MEDLINE | ID: mdl-16427862

BACKGROUND: Thrombin generated during cardiopulmonary bypass activates the high-affinity thrombin receptor, protease-activated receptor 1 (PAR1), causing platelet dysfunction and excessive bleeding. The serine protease inhibitor aprotinin protects platelets against thrombin-mediated PAR1 activation in vitro and in vivo. Here we have investigated three novel recombinant aprotinin variants with specific modifications to the active site lysine at amino acid position 15 (arginine-15, arginine-15-alanine-17, and valine-15-leucine-17) for their effect on PAR1-mediated platelet aggregation in vitro. METHODS: Aggregation studies were carried out using washed human platelets (n = 9) or platelet rich plasma (n = 7) from healthy volunteers activated with 1 or 5 nM thrombin. Recombinant aprotinin variants were used at the molar equivalent to 50 KIU/mL of the parent compound. The PAR1-specific antagonist peptide, FLLRN, was used at 500 microM. RESULTS: Platelet aggregation at low concentrations of thrombin (1 nM) was mediated exclusively through PAR1, as shown by inhibition of aggregation in the presence of FLLRN. At 1 nM thrombin, the mean percentage +/- SD aggregation of washed platelets was 68.6% +/- 12.3%. This was suppressed by each aprotinin variant at the 50 KIU/mL equivalent dose: arginine-15 (23.0% +/- 17.5%, p < 0.001); arginine-15-alanine-17 (33.3% +/- 22.9%, p < 0.01); aprotinin (37.5% +/- 19.4%, p < 0.05); valine-15-leucine-17 (50.0% +/- 16.1%, not significant)). At 5 nM thrombin, which activates both high (PAR1) and low-affinity (PAR4) thrombin receptors on platelets, FLLRN and aprotinin failed to block aggregation: this finding indicates that aprotinin selectively targeted PAR1. In platelet-rich plasma, aggregation at 1 nM thrombin was 77.1% +/- 10.0%, and this was inhibited in the following order: arginine-15 (30.1% +/- 9.6%, p < 0.001); arginine-15-alanine-17 (52.3% +/- 9.7%, p > 0.001); aprotinin (55.9% +/- 6.2%, p > 0.001); valine-15-leucine-17 (73.7% +/- 7.1%, not significant). CONCLUSIONS: Aprotinin variants differentially inhibit PAR1-mediated platelet aggregation. With more understanding of the mechanisms of action of aprotinin and its derivatives, safer and more efficacious aprotinin variants may become available for clinical use.


Aprotinin/pharmacology , Platelet Aggregation/drug effects , Receptor, PAR-1/drug effects , Serine Proteinase Inhibitors/pharmacology , Cardiopulmonary Bypass , Humans , In Vitro Techniques , Recombinant Proteins/pharmacology
8.
Ann Thorac Surg ; 75(5): 1648-50, 2003 May.
Article En | MEDLINE | ID: mdl-12735602

Castleman's disease presents as a peculiar type of lymph node hyperplasia. Traditionally, the disease has been classified on clinical grounds (solitary or multicentric) and by histologic appearance (hyaline vascular pattern, plasma cell predominance, or mixed lesions). It is now increasingly clear that there are different etiologies for each of these different subtypes. Reported associations include POEMS syndrome (polyneuropathy, organomegally, endocrinopathy, monoclonal gammopathy, and skin changes), paraneoplastic pemphigus, Hodgkin's disease, and follicular dendritic cell sarcoma. We present a case of Castleman's disease associated with myasthenia gravis, the third reported case in the literature. We discuss Castleman's disease and review the literature.


Castleman Disease/complications , Myasthenia Gravis/complications , Adult , Castleman Disease/pathology , Castleman Disease/surgery , Female , Humans , Lymph Nodes/pathology , Myasthenia Gravis/pathology , Myasthenia Gravis/surgery , Thymus Gland/pathology
9.
Ann Thorac Surg ; 74(5): 1698-700, 2002 Nov.
Article En | MEDLINE | ID: mdl-12440638

Surgeons are increasingly faced with patients suffering from complicated pathology in multiple organ systems, to which multiple therapeutic agents with complex adverse effects are often prescribed. We face a daily challenge in maintaining an up-to-date knowledge of these complications. Heparin is widely used in surgical practice, yet our awareness of its adverse effects, other than bleeding and thrombocytopenia, remains poor. We will present an example of heparin-induced hyperkalemia following administration for cardiopulmonary bypass and intraaortic balloon pump prophylaxis. This is a rare but serious complication of heparin therapy, not usually reported in the context of a cardiac surgical patient. We will also discuss the renal physiology leading to hyperkalemia and the options available for its management.


Angina, Unstable/surgery , Coronary Artery Bypass , Heparin/adverse effects , Hyperkalemia/chemically induced , Myocardial Infarction/surgery , Critical Care , Female , Heparin/administration & dosage , Humans , Hyperaldosteronism/blood , Hyperaldosteronism/chemically induced , Hyperkalemia/blood , Intra-Aortic Balloon Pumping , Kidney Function Tests , Middle Aged , Postoperative Complications/blood , Postoperative Complications/chemically induced , Potassium/blood
...