Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Pharmacokinet ; 62(6): 881-890, 2023 06.
Article in English | MEDLINE | ID: mdl-37140727

ABSTRACT

BACKGROUND AND OBJECTIVE: Cotadutide is a balanced glucagon-like peptide-1 and glucagon receptor dual agonist under development for the treatment of non-alcoholic steatohepatitis and type 2 diabetes with chronic kidney disease. We evaluated the pharmacokinetics (PK), safety and immunogenicity of a single dose of cotadutide in individuals with varying degrees of renal impairment. METHODS: In this phase I bridging study, individuals 18-85 years of age, with a body mass index of 17-40 kg/m2 and varying degrees of renal function {end-stage renal disease (ESRD; creatinine clearance [CrCl] < 20 mL/min); severe renal impairment (CrCl ≥ 20 to < 30 mL/min); lower moderate renal impairment (CrCl ≥ 30 to < 44 mL/min); upper moderate renal impairment (CrCl ≥ 45 to < 60 mL/min); normal renal function (CrCl ≥ 90 mL/min)} were treated with a single dose of subcutaneous cotadutide 100 µg under fasted conditions in the lower abdomen. The co-primary endpoints were area under the plasma concentration-time curve from time zero to 48 h (AUC48) and the maximum observed plasma concentration (Cmax) for cotadutide. Safety and immunogenicity were secondary endpoints. This trial is registered with ClinicalTrials.gov (NCT03235375). RESULTS: A total of 37 individuals were enrolled in the study (only three enrolled in the ESRD group, therefore this group was excluded from the primary PK analysis). AUC48 and Cmax values for cotadutide were similar across all renal function groups {severe renal impairment vs. normal renal function: AUC48 geometric mean ratio (GMR) 0.99 (90% confidence interval [CI] 0.76-1.29); lower moderate renal impairment versus normal renal function: AUC48 GMR 1.01 (90% CI 0.79-1.30); upper moderate renal impairment versus normal renal function: AUC48 GMR 1.09 (90% CI 0.82-1.43)}. A sensitivity analysis that combined the ESRD and severe renal impairment groups did not show notable changes in the AUC48 and Cmax GMRs. The incidences of treatment-emergent adverse events (TEAE) ranged from 42.9 to 72.7% across all groups and were mostly mild to moderate in severity. Only one patient had a grade III or worse TEAE during the study period. No positive antidrug antibody results were observed. CONCLUSIONS: These results suggest that the PK and tolerability of cotadutide are unaffected by renal function and that dose adjustments may not be required in individuals with renal impairment.


Subject(s)
Diabetes Mellitus, Type 2 , Kidney Failure, Chronic , Renal Insufficiency , Humans , Receptors, Glucagon/therapeutic use , Glucagon/therapeutic use , Glucagon-Like Peptide 1/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Renal Insufficiency/complications , Kidney Failure, Chronic/complications , Area Under Curve
2.
Diabetes Obes Metab ; 24(7): 1360-1369, 2022 07.
Article in English | MEDLINE | ID: mdl-35403793

ABSTRACT

AIM: To assess the efficacy, safety and tolerability of cotadutide in patients with type 2 diabetes mellitus and chronic kidney disease. MATERIALS AND METHODS: In this phase 2a study (NCT03550378), patients with body mass index 25-45 kg/m2 , estimated glomerular filtration rate 30-59 ml/min/1.73 m2 and type 2 diabetes [glycated haemoglobin 6.5-10.5% (48-91 mmol/mol)] controlled with insulin and/or oral therapy combination, were randomized 1:1 to once-daily subcutaneous cotadutide (50-300 µg) or placebo for 32 days. The primary endpoint was plasma glucose concentration assessed using a mixed-meal tolerance test. RESULTS: Participants receiving cotadutide (n = 21) had significant reductions in the mixed-meal tolerance test area under the glucose concentration-time curve (-26.71% vs. +3.68%, p < .001), more time in target glucose range on continuous glucose monitoring (+14.79% vs. -21.23%, p = .001) and significant reductions in absolute bodyweight (-3.41 kg vs. -0.13 kg, p < .001) versus placebo (n = 20). In patients with baseline micro- or macroalbuminuria (n = 18), urinary albumin-to-creatinine ratios decreased by 51% at day 32 with cotadutide versus placebo (p = .0504). No statistically significant difference was observed in mean change in estimated glomerular filtration rate between treatments. Mild/moderate adverse events occurred in 71.4% of participants receiving cotadutide and 35.0% receiving placebo. CONCLUSIONS: We established the efficacy of cotadutide in this patient population, with significantly improved postprandial glucose control and reduced bodyweight versus placebo. Reductions in urinary albumin-to-creatinine ratios suggest potential benefits of cotadutide on kidney function, supporting further evaluation in larger, longer-term clinical trials.


Subject(s)
Diabetes Mellitus, Type 2 , Renal Insufficiency, Chronic , Albumins , Blood Glucose , Blood Glucose Self-Monitoring , Body Weight , Creatinine , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Double-Blind Method , Glucagon/therapeutic use , Glucagon-Like Peptide 1/therapeutic use , Glucagon-Like Peptide-1 Receptor/agonists , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/adverse effects , Peptides , Receptors, Glucagon , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy
3.
Australas J Ageing ; 37(3): 184-193, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30022583

ABSTRACT

OBJECTIVES: There are growing concerns that social isolation presents risks to older people's health and well-being. Thus, the objective of the review was to explore how technology is currently being utilised to combat social isolation and increase social participation, hence improving social outcomes for older people. METHODS: A systematic review of the literature was conducted across the social science and human-computer interaction databases. RESULTS: A total of 36 papers met the inclusion criteria and were analysed using a four-step process. Findings were threefold, suggesting that: (i) technologies principally utilised social network services and touch-screen technologies; (ii) social outcomes are often ill-defined or not defined at all; and (iii) methodologies used to evaluate interventions were often limited and small-scale. CONCLUSION: Results suggest a need for studies that examine new and innovative forms of technology, evaluated with rigorous methodologies, and drawing on clear definitions about how these technologies address social isolation/participation.


Subject(s)
Aging/psychology , Computers, Handheld , Social Isolation , Social Networking , Social Participation , Age Factors , Aged , Attitude to Computers , Family Relations , Female , Friends , Humans , Internet , Male , Mobile Applications , Quality of Life , User-Computer Interface , Video Games
4.
J Biol Chem ; 282(32): 23306-15, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17565999

ABSTRACT

The regulation of PTEN intrinsic biochemical properties has not been fully elucidated. In this report, we investigated the role of the PTEN carboxyl-terminal tail domain in regulating its membrane targeting and catalytic functions. Characterization of a panel of PTEN phosphorylation site mutants revealed that mutating Ser-385 to alanine (S385A) promoted membrane localization in vivo and phosphatase activity in vitro. Furthermore, S385A mutation was associated with a substantial reduction in the phosphorylation of the Ser-380/Thr-382/Thr-383 cluster. Therefore, Ser-385 could prime additional dephosphorylation events to regulate PTEN catalytic activity. Moreover, substituting Ser-380/Thr-382/Thr-383 to phosphomimic residues reversed the phosphatase activity of the S385A mutation. Next, we further defined the underlying mechanisms responsible for the COOH-terminal tail region in modulating PTEN biological activity. We have identified an interaction between the 71-amino acid carboxyl-terminal tail region and the CBRIII motif of the C2 domain, which has been implicated in membrane binding. In addition, a synthetic phosphomimic peptide encompassing the phosphorylation site cluster between amino acids 368 and 390 within the tail region mediated the suppression of PTEN catalytic activity in vitro. This same peptide when expressed in cultured cells also impeded PTEN membrane localization and enhanced phospho-Akt levels. Thus, our data suggest that the COOH-terminal tail can act as an autoinhibitory domain to control both PTEN membrane recruitment and phosphatase activity.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/physiology , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Dogs , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Subcellular Fractions/metabolism
5.
Mol Cell Biol ; 26(19): 7145-54, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16980617

ABSTRACT

R-Ras3/M-Ras is a member of the RAS superfamily of small-molecular-weight GTP-binding proteins. Previous studies have demonstrated high levels of expression in several regions of the central nervous system, and a constitutively active form of M-Ras promotes cytoskeletal reorganization, cellular transformation, survival, and differentiation. However, the physiological functions of M-Ras during embryogenesis and postnatal development have not been elucidated. By using a specific M-Ras antibody, we demonstrated a high level of M-Ras expression in astrocytes, in addition to neurons. Endogenous M-Ras was activated by several trophic factors in astrocytes, including epidermal growth factor (EGF), basic fibroblast growth factor, and hepatocyte growth factor. Interestingly, M-Ras activation by EGF was more sustained compared to prototypic Ras. A mouse strain deficient in M-Ras was generated to investigate its role in development. M-Ras null mice appeared phenotypically normal, and there was a lack of detectable morphological and neurological defects. In addition, primary astrocytes derived from Mras(-/-) mice did not appear to display substantial alterations in the activation of both the mitogen-activated protein kinase and phosphatidylinositol 3-kinase pathways in response to trophic factors.


Subject(s)
Growth Substances/pharmacology , Monomeric GTP-Binding Proteins/metabolism , Signal Transduction/drug effects , Animals , Astrocytes/cytology , Astrocytes/drug effects , Enzyme Activation/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Monomeric GTP-Binding Proteins/deficiency , Monomeric GTP-Binding Proteins/genetics , Phosphotransferases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , ras Proteins
SELECTION OF CITATIONS
SEARCH DETAIL