Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Comput Methods Programs Biomed ; 244: 107968, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38064957

ABSTRACT

Pramlintide, an amylin analog, has been coming up as an agent in type 1 diabetes dual-hormone therapies (insulin/pramlintide). Since pramlintide slows down gastric emptying, it allows for easing glucose control and reducing the burden of meal announcements. Pre-clinical in silico evaluations are a key step in the development of any closed-loop strategy. However, mathematical models are needed, and pramlintide models in the literature are scarce. This work proposes a proof-of-concept pramlintide model, describing its subcutaneous pharmacokinetics (PK) and its effect on gastric emptying (PD). The model is validated with published populational (clinical) data. The model development is divided into three stages: intravenous PK, subcutaneous PK, and PD modeling. In each stage, a set of model structures are proposed, and their performance is assessed using the Akaike Information Criterion (AIC) and the Bayesian Information Criterion (BIC). In order to evaluate the modulation of the rate of gastric emptying, a literature meal model was used. The final pramlintide model comprises four compartments and a function that modulates gastric emptying depending on plasma pramlintide. Results show an appropriate fit for the data. Some aspects are left as open questions due to the lack of specific data (e.g., the influence of meal composition on the pramlintide effect). Moreover, further validation with individual data is necessary to propose a virtual cohort of patients.


Subject(s)
Diabetes Mellitus, Type 1 , Islet Amyloid Polypeptide , Humans , Islet Amyloid Polypeptide/pharmacokinetics , Islet Amyloid Polypeptide/therapeutic use , Hypoglycemic Agents/pharmacokinetics , Gastric Emptying , Bayes Theorem , Diabetes Mellitus, Type 1/drug therapy , Insulin , Blood Glucose
2.
Adv Sci (Weinh) ; 8(21): e2101575, 2021 11.
Article in English | MEDLINE | ID: mdl-34499434

ABSTRACT

Dual-hormone replacement therapy with insulin and amylin in patients with type 1 diabetes has the potential to improve glucose management. Unfortunately, currently available formulations require burdensome separate injections at mealtimes and have disparate pharmacokinetics that do not mimic endogenous co-secretion. Here, amphiphilic acrylamide copolymers are used to create a stable co-formulation of monomeric insulin and amylin analogues (lispro and pramlintide) with synchronous pharmacokinetics and ultra-rapid action. The co-formulation is stable for over 16 h under stressed aging conditions, whereas commercial insulin lispro (Humalog) aggregates in 8 h. The faster pharmacokinetics of monomeric insulin in this co-formulation result in increased insulin-pramlintide overlap of 75 ± 6% compared to only 47 ± 7% for separate injections. The co-formulation results in similar delay in gastric emptying compared to pramlintide delivered separately. In a glucose challenge, in rats, the co-formulation reduces deviation from baseline glucose compared to insulin only, or separate insulin and pramlintide administrations. Further, comparison of interspecies pharmacokinetics of monomeric pramlintide suggests that pharmacokinetics observed for the co-formulation will be well preserved in future translation to humans. Together these results suggest that the co-formulation has the potential to improve mealtime glucose management and reduce patient burden in the treatment of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Acetaminophen/chemistry , Acetaminophen/metabolism , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/pathology , Drug Compounding , Gastric Emptying , Glucose Tolerance Test , Half-Life , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacokinetics , Infusions, Subcutaneous , Insulin/analogs & derivatives , Insulin/pharmacokinetics , Insulin Lispro/pharmacokinetics , Insulin Lispro/therapeutic use , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/pharmacokinetics , Islet Amyloid Polypeptide/therapeutic use , Male , Rats , Rats, Sprague-Dawley
3.
Lancet ; 397(10286): 1736-1748, 2021 05 08.
Article in English | MEDLINE | ID: mdl-33894838

ABSTRACT

BACKGROUND: Cagrilintide, a long-acting amylin analogue, and semaglutide 2·4 mg, a glucagon-like peptide-1 analogue, are both being investigated as options for weight management. We aimed to determine the safety, tolerability, pharmacokinetics, and pharmacodynamics of this drug combination. METHODS: In this randomised, placebo-controlled, multiple-ascending dose, phase 1b trial, individuals aged 18-55 years with a body-mass index 27·0-39·9 kg/m2 and who were otherwise healthy were recruited from a single centre in the USA. The trial included six sequential overlapping cohorts, and in each cohort eligible participants were randomly assigned (3:1) to once-weekly subcutaneous cagrilintide (0·16, 0·30, 0·60, 1·2, 2·4, or 4·5 mg) or matched placebo, in combination with once-weekly subcutaneous semaglutide 2·4 mg, without lifestyle interventions. In each cohort, the doses of cagrilintide and semaglutide were co-escalated in 4-week intervals to the desired dose over 16 weeks, participants were treated at the target dose for 4 weeks, and then followed up for 5 weeks. Participants, investigators, and the sponsor were masked to treatment assignment. The primary endpoint was number of treatment-emergent adverse events from baseline to end of follow-up. Secondary pharmacokinetic endpoints assessed from day of last dose (week 19) to end of treatment (week 20) were area under the plasma concentration-time curve from 0 to 168 h (AUC0-168 h) and maximum concentration [Cmax] of cagrilintide and semaglutide; exploratory pharmacokinetic endpoints were half-life, time to Cmax [tmax], plasma clearance, and volume of distribution of cagrilintide and semaglutide; and exploratory pharmacodynamic endpoints were changes in bodyweight, glycaemic parameters, and hormones. Safety, pharmacokinetic, and pharmacodynamic endpoints were assessed in all participants who were exposed to at least one dose of study drug. This study is registered with ClinicalTrials.gov, NCT03600480, and is now complete. FINDINGS: Between July 25, 2018, and Dec 17, 2019, 285 individuals were screened and 96 were randomly assigned to cagrilintide (0·16-2·4 mg group n=12; 4·5 mg group n=11) or placebo (n=24), in combination with semaglutide 2·4 mg, of whom 95 were exposed to treatment (one patient in 0·60 mg cagrilintide group was not exposed) and included in the safety and full analysis datasets. The mean age was 40·6 years (SD 9·2), 56 (59%) of 95 participants were men and 51 (54%) were Black or African American. Of 566 adverse events reported in 92 participants (69 [97%] of 71 participants assigned to 0·16-4·5 mg cagrilintide and 23 [96%] of 24 assigned to placebo), 207 (37%) were gastrointestinal disorders. Most adverse events were mild to moderate in severity and the proportion of participants with one or more adverse event was similar across treatment groups. Exposure was proportional to cagrilintide dose and did not affect semaglutide exposure or elimination. AUC0-168 h ranged from 926 nmol × h/L to 24 271 nmol × h/L, and Cmax ranged from 6·14 nmol/L to 170 nmol/L with cagrilintide 0·16-4·5 mg. AUC0-168 h ranged from 12 757 nmol × h/L to 15 305 nmol × h/L, and Cmax ranged from 96·4 nmol/L to 120 nmol/L with semaglutide 2·4 mg. Cagrilintide 0·16-4·5 mg had a half-life of 159-195 h, with a median tmax of 24-72 h. Semaglutide 2·4 mg had a half-life of 145-165 h, with a median tmax of 12-24 h. Plasma clearance and volume of distribution for both cagrilintide and semaglutide were similar across treatment groups. At week 20, mean percentage bodyweight reductions were greater with cagrilintide 1·2 and 2·4 mg than with placebo (15·7% [SE 1·6] for cagrilintide 1·2 mg and 17·1% [1·5] for cagrilintide 2·4 mg vs 9·8% [1·2] for pooled placebo cohorts 1-5; estimated treatment difference of -6·0% [95% CI -9·9 to -2·0] for cagrilintide 1·2 mg and -7·4% [-11·2 to -3·5] for cagrilintide 2·4 mg vs pooled placebo), and with cagrilintide 4·5 mg than with matched placebo (15·4% [1·3] vs 8·0% [2·2]; estimated treatment difference -7·4% [-12·8 to -2·1]), all in combination with semaglutide 2·4 mg. Glycaemic parameters improved in all treatment groups, independently of cagrilintide dose. Changes in hormones were similar across treatment groups. INTERPRETATION: Concomitant treatment with cagrilintide and semaglutide 2·4 mg was well tolerated with an acceptable safety profile. Future larger and longer trials are needed to fully assess the efficacy and safety of this treatment combination. FUNDING: Novo Nordisk A/S.


Subject(s)
Anti-Obesity Agents/administration & dosage , Glucagon-Like Peptides/administration & dosage , Islet Amyloid Polypeptide/administration & dosage , Obesity/drug therapy , Weight Loss/drug effects , Adult , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Double-Blind Method , Drug Therapy, Combination , Female , Glucagon-Like Peptides/pharmacokinetics , Glucagon-Like Peptides/pharmacology , Humans , Injections , Islet Amyloid Polypeptide/adverse effects , Islet Amyloid Polypeptide/pharmacokinetics , Male , Middle Aged
4.
Nat Biomed Eng ; 4(5): 507-517, 2020 05.
Article in English | MEDLINE | ID: mdl-32393892

ABSTRACT

Treatment of patients with diabetes with insulin and pramlintide (an amylin analogue) is more effective than treatment with insulin only. However, because mixtures of insulin and pramlintide are unstable and have to be injected separately, amylin analogues are only used by 1.5% of people with diabetes needing rapid-acting insulin. Here, we show that the supramolecular modification of insulin and pramlintide with cucurbit[7]uril-conjugated polyethylene glycol improves the pharmacokinetics of the dual-hormone therapy and enhances postprandial glucagon suppression in diabetic pigs. The co-formulation is stable for over 100 h at 37 °C under continuous agitation, whereas commercial formulations of insulin analogues aggregate after 10 h under similar conditions. In diabetic rats, the administration of the stabilized co-formulation increased the area-of-overlap ratio of the pharmacokinetic curves of pramlintide and insulin from 0.4 ± 0.2 to 0.7 ± 0.1 (mean ± s.d.) for the separate administration of the hormones. The co-administration of supramolecularly stabilized insulin and pramlintide better mimics the endogenous kinetics of co-secreted insulin and amylin, and holds promise as a dual-hormone replacement therapy.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Drug Compounding , Glucagon/metabolism , Insulin/therapeutic use , Islet Amyloid Polypeptide/therapeutic use , Animals , Bridged-Ring Compounds/chemistry , Diffusion , Drug Administration Routes , Drug Stability , Hydrogen-Ion Concentration , Imidazoles/chemistry , Insulin/administration & dosage , Insulin/pharmacokinetics , Insulin/pharmacology , Islet Amyloid Polypeptide/administration & dosage , Islet Amyloid Polypeptide/pharmacokinetics , Islet Amyloid Polypeptide/pharmacology , Male , Polyethylene Glycols/chemistry , Rats, Sprague-Dawley , Signal Transduction/drug effects , Swine
5.
J Pharmacokinet Pharmacodyn ; 45(2): 215-233, 2018 04.
Article in English | MEDLINE | ID: mdl-29170989

ABSTRACT

The inhibitory effect of anti-obesity drugs on energy intake (EI) is counter-acted by feedback regulation of the appetite control circuit leading to drug tolerance. This complicates the design and interpretation of EI studies in rodents that are used for anti-obesity drug development. Here, we investigated a synthetic long-acting analogue of the appetite-suppressing peptide hormone amylin (LAMY) in lean and diet-induced obese (DIO) rats. EI and body weight (BW) were measured daily and LAMY concentrations in plasma were assessed using defined time points following subcutaneous administration of the LAMY at different dosing regimens. Overall, 6 pharmacodynamic (PD) studies including a total of 173 rats were considered in this evaluation. Treatment caused a dose-dependent reduction in EI and BW, although multiple dosing indicated the development of tolerance over time. This behavior could be adequately described by a population model including homeostatic feedback of EI and a turnover model describing the relationship between EI and BW. The model was evaluated by testing its ability to predict BW loss in a toxicology study and was utilized to improve the understanding of dosing regimens for obesity therapy. As such, the model proved to be a valuable tool for the design and interpretation of rodent studies used in anti-obesity drug development.


Subject(s)
Body Weight/drug effects , Energy Intake/drug effects , Islet Amyloid Polypeptide/pharmacology , Islet Amyloid Polypeptide/pharmacokinetics , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Diet, High-Fat/methods , Female , Male , Obesity/drug therapy , Obesity/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar
6.
J Diabetes Sci Technol ; 8(3): 529-42, 2014 May.
Article in English | MEDLINE | ID: mdl-24876617

ABSTRACT

Type 1 diabetes mellitus (T1DM) complications are significantly reduced when normoglycemic levels are maintained via intensive therapy. The artificial pancreas is designed for intensive glycemic control; however, large postprandial excursions after a meal result in poor glucose regulation. Pramlintide, a synthetic analog of the hormone amylin, reduces the severity of postprandial excursions by reducing appetite, suppressing glucagon release, and slowing the rate of gastric emptying. The goal of this study is to create a glucose-insulin-pramlintide physiological model that can be employed into a controller to improve current control approaches used in the artificial pancreas. A model of subcutaneous (SC) pramlintide pharmacokinetics (PK) was developed by revising an intravenous (IV) pramlintide PK model and adapting SC insulin PK from a glucose-insulin model. Gray-box modeling and least squares optimization were used to obtain parameter estimates. Pharmacodynamics (PD) were obtained by choosing parameters most applicable to pramlintide mechanisms and then testing using a proportional PD effect using least squares optimization. The model was fit and validated using 27 data sets, which included placebo, PK, and PD data. SC pramlintide PK root mean square error values range from 1.98 to 10.66 pmol/L. Pramlintide PD RMSE values range from 10.48 to 42.76 mg/dL. A new in silico model of the glucose-insulin-pramlintide regulatory system is presented. This model can be used as a platform to optimize dosing of both pramlintide and insulin as a combined therapy for glycemic regulation, and in the development of an artificial pancreas as the kernel for a model-based controller.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/pharmacokinetics , Insulin Lispro/pharmacokinetics , Islet Amyloid Polypeptide/pharmacokinetics , Models, Biological , Administration, Intravenous , Biomarkers/blood , Blood Glucose/metabolism , Computer Simulation , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/diagnosis , Eating , Humans , Hypoglycemic Agents/administration & dosage , Injections, Subcutaneous , Insulin Lispro/administration & dosage , Islet Amyloid Polypeptide/administration & dosage , Nonlinear Dynamics , Reproducibility of Results , Treatment Outcome
7.
AAPS J ; 15(1): 15-29, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23054970

ABSTRACT

This report describes a pharmacokinetic/pharmacodynamic model for pramlintide, an amylinomimetic, in type 1 diabetes mellitus (T1DM). Plasma glucose and drug concentrations were obtained following bolus and 2-h intravenous infusions of pramlintide at three dose levels or placebo in 25 T1DM subjects during the postprandial period in a crossover study. The original clinical data were reanalyzed by mechanism-based population modeling. Pramlintide pharmacokinetics followed a two-compartment model with zero-order infusion and first-order elimination. Pramlintide lowered overall postprandial plasma glucose AUC (AUC(net)) and delayed the time to peak plasma glucose after a meal (T (max)). The delay in glucose T (max) and reduction of AUC(net) indicate that overall plasma glucose concentrations might be affected by differing mechanisms of action of pramlintide. The observed increase in glucose T (max) following pramlintide treatment was independent of dose within the studied dose range and was adequately described by a dose-independent, maximum pramlintide effect on gastric emptying of glucose in the model. The inhibition of endogenous glucose production by pramlintide was described using a sigmoidal function with capacity and sensitivity parameter estimates of 0.995 for I (max) and 23.8 pmol/L for IC(50). The parameter estimates are in good agreement with literature values and the IC(50) is well within the range of postprandial plasma amylin concentrations in healthy humans, indicating physiological relevance of the pramlintide effect on glucagon secretion in the postprandial state. This model may prove to be useful in future clinical studies of other amylinomimetics or antidiabetic drugs with similar mechanisms of action.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/pharmacokinetics , Islet Amyloid Polypeptide/pharmacokinetics , Adult , Diabetes Mellitus, Type 1/metabolism , Humans , Islet Amyloid Polypeptide/pharmacology , Male , Models, Biological , Single-Blind Method
8.
Colloids Surf B Biointerfaces ; 94: 101-6, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22326343

ABSTRACT

Since its discovery the therapeutic use of the pancreatic hormone amylin has been limited due to its poor water solubility and propensity for amyloid aggregation. We have entrapped the human amylin protein in polymeric nanoparticles, using a single emulsion-solvent evaporation method and investigated its effectiveness in the controlled release of the peptide. Typical preparations composed of poly-ε-caprolactone had a mean particle size of approximately 200 nm, low polydispersity index, high protein entrapment efficiency (80%) and process yield (90%), and spherical and smooth surfaces. These nanoparticles presented a controlled release in vitro for approximately 240 h. Pharmacological evaluation in vivo by subcutaneous administration in fasting mice demonstrated the bioactivity and effectiveness of the released human amylin, resulting in reduced glycemia lasting for at least 36 h. These features indicate the potential for the use of a confined particulate system in the therapeutic controlled and sustained release of human amylin.


Subject(s)
Appetite Depressants/pharmacokinetics , Delayed-Action Preparations/chemical synthesis , Islet Amyloid Polypeptide/pharmacokinetics , Nanoparticles/chemistry , Polyesters/chemistry , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/chemistry , Blood Glucose/analysis , Blood Glucose/drug effects , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Compounding , Emulsions , Fasting , Humans , Injections, Subcutaneous , Islet Amyloid Polypeptide/administration & dosage , Islet Amyloid Polypeptide/chemistry , Male , Mice , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Particle Size , Solubility
9.
FASEB J ; 26(3): 1228-38, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22183778

ABSTRACT

Islet amyloid polypeptide (IAPP) is a peptide hormone cosecreted with insulin by pancreatic ß-cells. In type II diabetes, IAPP aggregates in a process that is associated with ß-cell dysfunction and loss of ß-cell mass. The relationship between IAPP's conformational landscape and its capacity to mediate cell death remains poorly understood. We have addressed these unknowns by comparing the cytotoxic effects of sequence variants with differing α-helical and amyloid propensities. IAPP was previously shown to oligomerize cooperatively on binding to lipid bilayers. Here, comparable transitions are evident in cell culture and are associated with a change in subcellular localization to the mitochondria under toxic conditions. Notably, we find that this toxic gain of function maps to IAPP's capacity to adopt aggregated membrane-bound α-helical, and not ß-sheet, states. Our findings suggest that upon α-helical mediated oligomerization, IAPP acquires cell-penetrating peptide (CPP) properties, facilitating access to the mitochondrial compartment, resulting in its dysfunction.


Subject(s)
Insulin-Secreting Cells/metabolism , Intracellular Space/metabolism , Islet Amyloid Polypeptide/pharmacokinetics , Amino Acid Sequence , Animals , COS Cells , Cell Line, Tumor , Cell Survival/drug effects , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Insulin-Secreting Cells/pathology , Insulinoma/metabolism , Insulinoma/pathology , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/pharmacology , Microscopy, Confocal , Mitochondria/enzymology , Mitochondria/metabolism , Molecular Sequence Data , Oxidoreductases/metabolism , Protein Multimerization , Protein Structure, Secondary , Rats , Sequence Homology, Amino Acid , Spectrometry, Fluorescence
10.
Expert Opin Pharmacother ; 12(9): 1439-51, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21564002

ABSTRACT

INTRODUCTION: Postprandial glucose excursions negatively affect glycemic control and markers of cardiovascular health. Pramlintide, an amylinomimetic, is approved for treatment of elevated postprandial glucose levels in type 1 and type 2 diabetes mellitus. AREAS COVERED: A literature search of PubMed was conducted to locate articles (up to January 2011) pertaining to original preclinical and clinical research and reviews of amylin and pramlintide. Additional sources were selected from reference lists within articles obtained through the original literature search and from the internet. This article describes the known effects of endogenous amylin and the pharmacodynamics, pharmacokinetics and clinical efficacy of pramlintide. Drug-drug interactions and safety and tolerability are also reviewed. EXPERT OPINION: Pramlintide significantly reduces hemoglobin A(1c) and body weight in patients with type 1 and type 2 diabetes mellitus. Newer research is focusing on weight loss effects of pramlintide and pramlintide plus metreleptin in nondiabetic obese individuals. Preliminary results of these studies are discussed.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Islet Amyloid Polypeptide/pharmacokinetics , Islet Amyloid Polypeptide/therapeutic use , Drug Interactions , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...