Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
1.
Diabetes Obes Metab ; 24(12): 2411-2419, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-35892256

RÉSUMÉ

AIM: To report the in vitro and in vivo preclinical pharmacokinetic (PK) and pharmacodynamic (PD) properties of RA15127343, a novel ultralong-acting insulin analogue targeting once-weekly administration, in female Göttingen minipigs. METHODS: In vitro binding and activation of human insulin receptor isoforms (IR-A/IR-B), glucose uptake in rat myocytes, as well as mitogenic activity of RA15127343 were evaluated. In vivo, the PK and PD activities of RA15127343 were assessed in female, normoglycaemic Göttingen minipigs. The half-life (t1/2 ) and time to maximum plasma concentration (Tmax ) of subcutaneously (SC) administered RA15127343 (10/30/45/60 nmol/kg) were estimated. In vivo blood glucose and endogenous plasma C-peptide concentrations after single SC administration (10/30/45/60 nmol/kg) or repeated dosing (15 nmol/kg) were analysed. RESULTS: In comparison to human insulin, RA15127343 showed lower in vitro binding affinity (19.9/6.31 µM vs. 1.10/1.14 nM) and activation (2.054 µM/669.6 nM vs. 26.04/18.24 nM) of IR-A/IR-B, lower potency to activate glucose uptake (855.2 vs. 3.37 nM) and lower mitogenic activity (17.92 µM vs. 10.78 nM; proliferation in MCF7 cells). In vivo, the mean t1/2 and Tmax of RA15127343 after SC administration ranged from 48 to 59 and 30 to 39 hours, respectively. Blood glucose and plasma C-peptide concentrations were significantly lower with RA15127343 (single/repeated doses) versus vehicle. CONCLUSIONS: RA15127343 showed an ultra-long t1/2 with a slow onset of action. The preclinical pharmacological outcomes suggest RA15127343 could be a potential ultralong-acting insulin analogue with low risk of hypoglycaemia in humans.


Sujet(s)
Glycémie , Hypoglycémiants , Animaux , Femelle , Suidae , Humains , Rats , Glycémie/métabolisme , Hypoglycémiants/pharmacologie , Hypoglycémiants/usage thérapeutique , Peptide C , Porc miniature/métabolisme , Insuline à longue durée d'action , Insuline/pharmacologie
2.
J Control Release ; 338: 8-21, 2021 10 10.
Article de Anglais | MEDLINE | ID: mdl-34298056

RÉSUMÉ

Delivery of therapeutic peptides upon oral administration is highly desired and investigations report that the cell-penetrating peptide (CPP) penetratin and its analogues shuffle and penetramax show potential as carriers to enhance insulin delivery. Exploring this, the specific aim of the present study was to understand the impact that their complexation with a lipidated or non-lipidated therapeutic cargo would have on the delivery, to evaluate the effect of differences in membrane interactions in vitro and in vivo, as well as to deduce the mode of action leading to enhanced delivery. Fundamental biophysical aspects were studied by a range of orthogonal methods. Transepithelial permeation of therapeutic peptide was evaluated using the Caco-2 cell culture model supplemented with epithelial integrity measurements, real-time assessment of the carrier peptide effects on cell viability and on mode of action. Pharmacokinetic and pharmacodynamic (PK/PD) parameters were evaluated following intestinal administration to rats and tissue effects were investigated by histology. The biophysical studies revealed complexation of insulin with shuffle and penetramax, but not with penetratin. This corresponded to enhanced transepithelial permeation of insulin, but not of lipidated insulin, when in physical mixture with shuffle or penetramax. The addition of shuffle and penetramax was associated with a lowering of Caco-2 cell monolayer integrity and viability, where the lowering of cell viability was immediate, but reversible. Insulin delivery in rats was enhanced by shuffle and penetramax and accompanied by a 10-20-fold decrease in blood glucose with immediate effect on the intestinal mucosa. In conclusion, shuffle and penetramax, but not penetratin, demonstrated to be potential candidates as carriers for transmucosal delivery of insulin upon oral administration, and their effect depended on association with both cargo and cell membrane. Interestingly, the present study provides novel mechanistic insight that peptide carrier-induced cargo permeation points towards enhancement via the paracellular route in the tight epithelium. This is different from the anticipated belief being that it is the cell-penetrating capability that facilitate transepithelial cargo permeation via a transcellular route.


Sujet(s)
Peptides de pénétration cellulaire , Insuline , Administration par voie orale , Animaux , Cellules Caco-2 , Protéines de transport , Peptides de pénétration cellulaire/métabolisme , Humains , Muqueuse intestinale/métabolisme , Rats
3.
Diabetes Obes Metab ; 23(1): 166-174, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33001554

RÉSUMÉ

AIMS: Insulin glargine 300 U/mL (Gla-300) contains the same units versus glargine 100 U/mL (Gla-100) in three-fold lower volume, and higher subcutaneous (SC) doses are required in people with diabetes. To investigate blood glucose (BG) lowering potency, Gla-300 and Gla-100 were compared after intravenous (IV, for 4 h) and SC (for 24 h) injection in healthy Beagle dogs. MATERIALS AND METHODS: The dose of 0.15 U/kg Gla-300 and Gla-100 was injected IV in 12 dogs. BG, C-peptide, glucagon and the active metabolite 21A-Gly-human insulin (M1; liquid chromatography-tandem mass spectrometry method) were measured. Twelve other dogs were studied after SC injection of 0.3 U/kg Gla-300 and Gla-100. RESULTS: After IV injection, Gla-300 and Gla-100 were equally potent [BG_AUC0-4 h ratio 1.01 (95% confidence interval, 0.94; 1.09)]. After SC injection, BG decreased slower and less with Gla-300. Similar metabolism of Gla-300 and Gla-100 to M1 occurred with IV dosing [M1_AUC0-1 h ratio 0.99 (95% confidence interval, 0.82; 1.22)], but with SC dosing M1_Cmax and AUC0-24h were 44% and 17% lower; mean residency time and bioavailability were 32% longer and 50% lower, with Gla-300. CONCLUSIONS: IV Gla-300 and Gla-100 have the equivalent of BG-lowering potency and M1 metabolism. SC Gla-300 has lower M1 bioavailability with a reduced BG-lowering effect and need for greater doses versus Gla-100.


Sujet(s)
Diabète de type 2 , Hypoglycémiants , Animaux , Biodisponibilité , Glycémie , Chiens , Insuline glargine , Insuline à longue durée d'action
4.
Protein Eng Des Sel ; 332020 09 14.
Article de Anglais | MEDLINE | ID: mdl-33159202

RÉSUMÉ

Insulin is a peptide hormone produced by the pancreas. The physiological role of insulin is the regulation of glucose metabolism. Under certain pathological conditions the insulin levels can be reduced leading to the metabolic disorder diabetes mellitus (DM). For type 1 DM and, dependent on the disease progression for type 2 DM, insulin substitution becomes indispensable. To relieve insulin substitution therapy for patients, novel insulin analogs with pharmacokinetic and pharmacodynamic profiles aiming for long-lasting or fast-acting insulins have been developed. The next step in the evolution of novel insulins should be insulin analogs with a time action profile beyond 1-2 days, preferable up to 1 week. Nowadays, insulin is produced in a recombinant manner. This approach facilitates the design and production of further insulin-analogs or insulin-fusion proteins. The usage of the Fc-domain from immunoglobulin as a fusion partner for therapeutic proteins and peptides is widely used to extend their plasma half-life. Insulin consists of two chains, the A- and B-chain, which are connected by two disulfide-bridges. To produce a novel kind of Fc-fusion protein we have fused the A-chain as well as the B-chain to Fc-fragments containing either 'knob' or 'hole' mutations. The 'knob-into-hole' technique is frequently used to force heterodimerization of the Fc-domain. Using this approach, we were able to produce different variants of two-chain-insulin-Fc-protein (tcI-Fc-protein) variants. The tcI-Fc-fusion variants retained activity as shown in in vitro assays. Finally, prolonged blood glucose lowering activity was demonstrated in normoglycemic rats. Overall, we describe here the production of novel insulin-Fc-fusion proteins with prolonged times of action.


Sujet(s)
Glycémie/métabolisme , Fragments Fc des immunoglobulines , Insuline , Protéines de fusion recombinantes , Animaux , Humains , Fragments Fc des immunoglobulines/biosynthèse , Fragments Fc des immunoglobulines/génétique , Fragments Fc des immunoglobulines/pharmacologie , Insuline/biosynthèse , Insuline/génétique , Insuline/pharmacologie , Mâle , Rats , Rat Sprague-Dawley , Protéines de fusion recombinantes/biosynthèse , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/pharmacologie
5.
Drug Dev Ind Pharm ; 46(7): 1073-1079, 2020 Jul.
Article de Anglais | MEDLINE | ID: mdl-32478645

RÉSUMÉ

Long-term stability is one of the main challenges for translation of therapeutic proteins into commercially viable biopharmaceutical products. During processing and storage, proteins are susceptible to denaturation. The aim of this work was to evaluate the stability of amphiphilic cyclodextrin-based nanoparticles (NPs) containing insulin glulisine. The stability of the NP dispersion was systematically evaluated following storage at three different temperatures (4 °C, room temperature (RT) and 40 °C). While the colloidal parameters of the NPs in terms of size and zeta potential were maintained (109 ± 9 nm, polydispersity index 0.272, negative zeta potential -25 ± 3 mV), insulin degraded over 60 days during storage. To enhance the shelf life of the product and to circumvent the need for cold-chain maintenance, a lyophilized formulation containing insulin glulisine NPs (1.75 mg/mL of NPs) and 25 mg/mL trehalose was produced. The freeze-dried powder extended the stability of the product for up to 30 days at ambient temperature and 90 days at 4 °C (with 95% and >80% insulin recovery, respectively). Following intra-intestinal administration of the freeze-dried formulation, while no lowering of blood glucose was seen, insulin glulisine was detected in both portal and systemic blood indicating that potential exists for further development of the formulation to simultaneously achieve prolonged stability and therapeutic efficacy.


Sujet(s)
Cyclodextrines , Insuline/analogues et dérivés , Nanoparticules , Stabilité de médicament , Lyophilisation , Insuline/composition chimique
6.
Front Pharmacol ; 11: 591293, 2020.
Article de Anglais | MEDLINE | ID: mdl-33679386

RÉSUMÉ

Background: Targeting long-lasting insulins to the liver may improve metabolic alterations that are not corrected with current insulin replacement therapies. However, insulin is only able to promote lipogenesis but not to block gluconeogenesis in the insulin-resistant liver, exacerbating liver steatosis associated with diabetes. Methods: In order to overcome this limitation, we fused a single-chain insulin to apolipoprotein A-I, and we evaluated the pharmacokinetics and pharmacodynamics of this novel fusion protein in wild type mice and in db/db mice using both recombinant proteins and recombinant adenoassociated virus (AAV). Results: Here, we report that the fusion protein between single-chain insulin and apolipoprotein A-I prolonged the insulin half-life in circulation, and accumulated in the liver. We analyzed the long-term effect of these insulin fused to apolipoprotein A-I or insulin fused to albumin using AAVs in the db/db mouse model of diabetes, obesity, and liver steatosis. While AAV encoding insulin fused to albumin exacerbated liver steatosis in several mice, AAV encoding insulin fused to apolipoprotein A-I reduced liver steatosis. These results were confirmed upon daily subcutaneous administration of the recombinant insulin-apolipoprotein A-I fusion protein for six weeks. The reduced liver steatosis was associated with reduced body weight in mice treated with insulin fused to apolipoprotein A-I. Recombinant apolipoprotein A-I alone significantly reduces body weight and liver weight, indicating that the apolipoprotein A-I moiety is the main driver of these effects. Conclusion: The fusion protein of insulin and apolipoprotein A-I could be a promising insulin derivative for the treatment of diabetic patients with associated fatty liver disease.

7.
ACS Appl Mater Interfaces ; 12(1): 1257-1269, 2020 Jan 08.
Article de Anglais | MEDLINE | ID: mdl-31802658

RÉSUMÉ

Nanoparticle constructs for oral peptide delivery at a minimum must protect and present the peptide at the small intestinal epithelium in order to achieve oral bioavailability. In a reproducible, scalable, surfactant-free process, a core was formed with insulin in ratios with two established excipients and stabilizers, zinc chloride and l-arginine. Cross-linking was achieved with silica, which formed an outer shell. The process was reproducible across several batches, and physicochemical characterization of a single batch was confirmed in two independent laboratories. The silica-coated nanoparticles (SiNPs) entrapped insulin with high entrapment efficiency, preserved its structure, and released it at a pH value present in the small intestine. The SiNP delivered insulin to the circulation and reduced plasma glucose in a rat jejunal instillation model. The delivery mechanism required residual l-arginine in the particle to act as a permeation enhancer for SiNP-released insulin in the jejunum. The synthetic process was varied in terms of ratios of zinc chloride and l-arginine in the core to entrap the glucagon-like peptide 1 analogue, exenatide, and bovine serum albumin. SiNP-delivered exenatide was also bioactive in mice to some extent following oral gavage. The process is the basis for a platform for oral peptide and protein delivery.


Sujet(s)
Arginine/composition chimique , Nanoparticules/composition chimique , Silice/composition chimique , Zinc/composition chimique , Administration par voie orale , Animaux , Chromatographie en phase liquide à haute performance , Dichroïsme circulaire , Femelle , Glucagon-like peptide 1/composition chimique , Spectroscopie par résonance magnétique , Souris , Souris de lignée C57BL , Peptides/composition chimique
8.
Int J Pharm ; 560: 377-384, 2019 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-30790612

RÉSUMÉ

To face the challenges of oral delivery of peptide and protein (P/P) drugs, self-emulsifying drug delivery systems (SEDDSs) containing monoacyl phosphatidylcholine (MAPC), Labrasol (LAB) and medium-chain (MC) monoglycerides as permeation enhancers (PEs) were evaluated for their effect on intestinal absorption of insulin. In this study, insulin was complexed with phosphatidylcholine (SPC) to form an insulin-SPC complex (ins-SPC) with increased lipophilicity. The following three SEDDSs: MCT(MAPC) (MC triglycerides and MAPC included), MCT(RH40) (MC triglycerides and Kolliphor® RH40 included) and LCT(MAPC) (long-chain triglycerides and MAPC included) were loading with ins-SPC (4% or 8% w/w of SPC). Three SEDDSs generated emulsions with droplet sizes between 50 and 470 nm and with zeta potentials between -5 to -25 mV in a simulated intestinal medium. Mucus-secreting Caco-2/HT29-MTX-E12 co-culture and Caco-2 monolayers were used as in vitro cell transport models to investigate insulin permeability. In comparison to insulin HBSS solution, MCT(MAPC) significantly increased the insulin permeability across co-culture and Caco-2 monolayers (2.0-2.5 × 10-7 cm/s). In an intra-jejunal (i.j.) instillation model in rats, MCT(RH40) significantly decreased the rat blood glucose after 0.5 h by 17.0 ±â€¯2.5% and for MCT(MAPC), it was 23.6 ±â€¯10.6%. Furthermore, a lipase inhibitor orlistat was incorporated into MCT(MAPC) to evaluate the effect of lipid digestion on insulin absorption. Results indicated that the incorporation of orlistat did not significantly alter the in vivo insulin absorption. Overall, the SEDDS MCT(MAPC) composed of natural PEs (MAPC and MC glycerides) and synthetic PE (LAB) significantly increased the intestinal absorption of insulin upon i.j. instillation. Although it is not possible to conclude if a single PE is dominating the intestinal absorption of insulin, MCT(MAPC) seems to have the potential for oral insulin delivery.


Sujet(s)
Systèmes de délivrance de médicaments , Excipients/composition chimique , Hypoglycémiants/administration et posologie , Insuline/administration et posologie , Animaux , Cellules Caco-2 , Techniques de coculture , Émulsions , Glycérides/composition chimique , Cellules HT29 , Humains , Hypoglycémiants/pharmacocinétique , Insuline/pharmacocinétique , Absorption intestinale , Jéjunum/métabolisme , Mâle , Modèles biologiques , Monoglycérides/composition chimique , Orlistat/administration et posologie , Orlistat/pharmacologie , Taille de particule , Perméabilité , Phosphatidylcholines/composition chimique , Rats , Rat Sprague-Dawley
9.
J Control Release ; 286: 402-414, 2018 09 28.
Article de Anglais | MEDLINE | ID: mdl-30075208

RÉSUMÉ

Due to excellent efficacy, low toxicity, and well-defined selectivity, development of new injectable peptides is increasing. However, the translation of these drugs into products for effective oral delivery has been restricted due to poor oral bioavailability. Nanoparticle (NP) formulations have potential to overcome the barriers to oral peptide delivery through protecting the payload and increasing bioavailability. This study describes the rational design, optimization and evaluation of a cyclodextrin-based NP entrapping insulin glulisine for intestinal administration. A cationic amphiphilic cyclodextrin (click propyl-amine cyclodextrin (CD)) was selected as the primary complexing agent for NP development. Following NP synthesis, in vitro characterization was performed. The insulin glulisine NPs exhibited an average size of 109 ±â€¯9 nm, low polydispersity index (0.272) negative zeta potential (-25 ±â€¯3 mV), high association efficiency (71.4 ±â€¯3.37%) and an insulin loading of 10.2%. In addition, the NPs exhibited colloidal stability in intestinal-biorelevant media (SIF, supplemented-SIF 1% (w/v) and FaSSIF-V2) for up to 4 h. Proteolysis studies indicated that the NPs conferred protection to the entrapped insulin relative to free insulin. In vivo rat jejunal instillation studies demonstrated that the NPs mediated systemic insulin absorption, accompanied by a decrease in blood glucose levels. The relative bioavailability of the instilled insulin (50 IU/kg) from the NP was 5.5% compared to subcutaneous administration of insulin solution (1 IU/kg). The pharmacodynamic and pharmacokinetic data indicate that this cyclodextrin-based formulation may have potential for further research as an oral insulin dosage form.


Sujet(s)
Cyclodextrines/composition chimique , Vecteurs de médicaments/composition chimique , Hypoglycémiants/administration et posologie , Insuline/analogues et dérivés , Nanoparticules/composition chimique , Tensioactifs/composition chimique , Administration par voie orale , Animaux , Biodisponibilité , Cellules HepG2 , Humains , Hypoglycémiants/composition chimique , Hypoglycémiants/pharmacocinétique , Hypoglycémiants/pharmacologie , Insuline/administration et posologie , Insuline/composition chimique , Insuline/pharmacocinétique , Insuline/pharmacologie , Absorption intestinale , Mâle , Modèles moléculaires , Stabilité protéique , Rat Wistar , Suidae
11.
Diabetologia ; 59(9): 2018-25, 2016 09.
Article de Anglais | MEDLINE | ID: mdl-27241182

RÉSUMÉ

AIMS/HYPOTHESIS: Previous epidemiological studies have reported a potential link between insulin analogues and breast cancer; however, a prospective randomised controlled trial showed neutral effects of insulin glargine on cancer risk. Insulin glargine is metabolised in vivo to an M1 metabolite. A question remains whether a subset of individuals with slower rates of glargine metabolism or who are on high doses could, theoretically, have an increased risk of cancer progression if a tumour is already present. In this study, we aimed to determine whether a non-metabolisable form of insulin glargine induced murine breast cancer growth. METHODS: A mouse model of type 2 diabetes (MKR) was used for these studies. MKR mice were injected with two murine mammary cancer cell lines: Mvt-1 cells (derived from MMTV-c-Myc/Vegf tumours) and Met1 cells (derived from MMTV-polyoma virus middle T antigen tumours). Mice were treated with 25 U/kg per day of the long-acting insulin analogues, insulin glargine, insulin detemir, insulin degludec or non-metabolisable glargine, or vehicle. RESULTS: No difference in tumour growth was seen in terms of tumour size after insulin glargine, detemir, degludec or vehicle injections. Non-metabolisable glargine did not increase tumour growth compared with insulin glargine or vehicle. Insulin glargine and non-metabolisable glargine led to insulin receptor phosphorylation in vivo rather than IGF-1 receptor phosphorylation. CONCLUSIONS/INTERPRETATION: These results demonstrate that in a mouse model of type 2 diabetes, at high concentrations, basal insulin analogues and a non-metabolisable glargine analogue do not promote the progression of breast tumours.


Sujet(s)
Diabète de type 2/traitement médicamenteux , Insuline glargine/effets indésirables , Tumeurs mammaires de l'animal/induit chimiquement , Tumeurs mammaires de l'animal/anatomopathologie , Animaux , Lignée cellulaire tumorale , Diabète de type 2/sang , Modèles animaux de maladie humaine , Femelle , Humains , Hypoglycémiants/effets indésirables , Hypoglycémiants/composition chimique , Hypoglycémiants/usage thérapeutique , Insuline/composition chimique , Insuline/usage thérapeutique , Insuline glargine/composition chimique , Insuline glargine/usage thérapeutique , Souris , Phosphorylation/effets des médicaments et des substances chimiques , Récepteur à l'insuline/métabolisme , Récepteurs des somatomédines/métabolisme
12.
Adv Drug Deliv Rev ; 106(Pt B): 196-222, 2016 11 15.
Article de Anglais | MEDLINE | ID: mdl-26964477

RÉSUMÉ

While some orally delivered diabetes peptides are moving to late development with standard formulations incorporating functional excipients, the demonstration of the value of nanotechnology in clinic is still at an early stage. The goal of this review is to compare these two drug delivery approaches from a physico-chemical and a biopharmaceutical standpoint in an attempt to define how nanotechnology-based products can be differentiated from standard oral dosage forms for oral bioavailability of diabetes peptides. Points to consider in a translational approach are outlined to seize the opportunities offered by a better understanding of both the intestinal barrier and of nano-carriers designed for oral delivery.


Sujet(s)
Diabète/métabolisme , Systèmes de délivrance de médicaments , Excipients/composition chimique , Nanomédecine , Peptides/administration et posologie , Peptides/pharmacocinétique , , Administration par voie orale , Animaux , Excipients/administration et posologie , Excipients/pharmacocinétique , Humains
13.
Arch Physiol Biochem ; 120(4): 158-65, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25144413

RÉSUMÉ

CONTEXT: Insulin glargine (GLA) is rapidly metabolized in vivo to metabolite M1, which has in vitro metabolic and mitogenic profiles comparable with human insulin (HI). OBJECTIVE: To investigate the pharmacologic and signalling profiles of a non-metabolizable analogue (A21Gly,DiD-Arg) insulin (D-GLA). METHODS: Rats were injected s.c. with 1, 12.5 or 200 U/kg of GLA or D-GLA; blood glucose and phosphorylation status of the insulin receptor (IR), Akt and IGF-1 receptor (IGF1R) in tissue samples were investigated after 1 h. Plasma samples were analysed for insulin by LC-MS/MS. RESULTS: Blood glucose lowering was prolonged with D-GLA. D-GLA comprised ≥98% of insulin after D-GLA injection; M1 comprised 76-92% after GLA injection. IR and Akt phosphorylation were comparable with GLA and D-GLA. Neither analogue stimulated IGF1R phosphorylation. CONCLUSIONS: Suprapharmacological doses of D-GLA did not activate IGF1R in vivo. Mitogenic effects of insulin and insulin analogues might be solely based on IR growth-promoting activity.


Sujet(s)
Glycémie/effets des médicaments et des substances chimiques , Hypoglycémiants/pharmacologie , Insuline à longue durée d'action/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Récepteur IGF de type 1/métabolisme , Récepteur à l'insuline/métabolisme , Animaux , Humains , Insuline glargine , Mâle , Phosphorylation/effets des médicaments et des substances chimiques , Rats , Rat Wistar , Transduction du signal/effets des médicaments et des substances chimiques , Spectrométrie de masse en tandem
14.
Expert Opin Drug Discov ; 9(10): 1223-51, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25119443

RÉSUMÉ

INTRODUCTION: Lixisenatide is a once-daily short-acting glucagon-like peptide-1 (GLP-1) receptor agonist (GLP-1RA) used in the treatment of type 2 diabetes mellitus (T2DM). It is used in combination with oral antidiabetics and/or basal insulin in patients inadequately controlled on these medications and who are undergoing diet and lifestyle modification. GLP-1RAs glucose-dependently increase insulin secretion, decrease glucagon secretion, and slow gastric emptying, thereby improving glycemic control. GLP-1RAs are associated with body weight benefits and low rates of hypoglycemia which are welcome in patients with T2DM. AREAS COVERED: The authors describe the identification of GLP-1RAs as suitable targets for modification with structure-inducing probe technology to improve stability and resistance to proteolytic degradation. Clinical studies have assessed lixisenatide across > 5000 patients as a monotherapy or add-on to a variety of commonly used antidiabetic medications. These studies highlighted the effects of lixisenatide on gastric emptying, explaining its particular improvements in postprandial plasma glucose (PPG) excursions and underscoring its efficacy in combination with insulin glargine. Lixisenatide was well tolerated, with nausea and vomiting being the most frequently reported adverse events. EXPERT OPINION: The once-daily administration of lixisenatide as well as its substantial sustained effect on gastric emptying and, hence, PPG excursions are all important features compared with the other GLP-1RAs. The combination of two injectables, such as basal insulin to lower fasting plasma glucose and a GLP-1RA that curtails PPG excursions, is clinically valuable and could differentiate lixisenatide from other GLP-1RAs, especially from those continuously acting GLP-1RAs with little effect on gastric emptying and PPG excursions.


Sujet(s)
Diabète expérimental/traitement médicamenteux , Diabète de type 2/traitement médicamenteux , Découverte de médicament/méthodes , Hypoglycémiants , Peptides , Récepteurs au glucagon/agonistes , Animaux , Glycémie/analyse , Essais cliniques comme sujet , Diabète expérimental/métabolisme , Diabète de type 2/métabolisme , Conception de médicament , Évaluation préclinique de médicament , Glucagon-like peptide 1/métabolisme , Récepteur du peptide-1 similaire au glucagon , Humains , Hypoglycémiants/administration et posologie , Hypoglycémiants/composition chimique , Hypoglycémiants/usage thérapeutique , Peptides/administration et posologie , Peptides/composition chimique , Peptides/usage thérapeutique , Liaison aux protéines , Résultat thérapeutique
15.
J Diabetes Complications ; 28(1): 110-4, 2014.
Article de Anglais | MEDLINE | ID: mdl-23992745

RÉSUMÉ

In addition to promoting glucose homeostasis, glucagon-like peptide 1 (GLP-1) has a number of extra-pancreatic effects that regulate appetite and body weight. GLP-1 delays gastric emptying, which is vital for postprandial glucose (PPG) control. As GLP-1 is rapidly degraded by protease dipeptidyl peptidase-4, a number of degradation-resistant GLP-1 receptor agonists (GLP-1RAs) have been developed for the treatment of Type 2 diabetes mellitus. These agents can be broadly categorized as being short- or long-acting, based on their pharmacokinetic profile. Short-acting agonists predominantly affect PPG and delay gastric emptying in a sustained manner, whereas long-acting agents largely affect fasting plasma glucose and their delay in gastric emptying appears to be subjected to tachyphylaxis. Lixisenatide is a "short-acting" once-daily prandial GLP-1RA. This review provides an overview of the preclinical studies that are currently available and that evaluate the efficacy of lixisenatide on gastric emptying and PPG levels. The preclinical evidence outlined in this review supports the efficacy of lixisenatide in reducing PPG excursions and delaying gastric emptying. Furthermore, in contrast to long-acting agents, the actions of lixisenatide do not appear to be subjected to tachyphylaxis.


Sujet(s)
Glycémie/effets des médicaments et des substances chimiques , Vidange gastrique/effets des médicaments et des substances chimiques , Peptides/pharmacologie , Période post-prandiale/effets des médicaments et des substances chimiques , Animaux , Glycémie/métabolisme , Essais cliniques comme sujet , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Diabète de type 2/physiopathologie , Évaluation préclinique de médicament/statistiques et données numériques , Récepteur du peptide-1 similaire au glucagon , Humains , Récepteurs au glucagon/agonistes , Résultat thérapeutique
16.
Am J Physiol Endocrinol Metab ; 305(12): E1473-82, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-24148347

RÉSUMÉ

The impact of the GLP-1 receptor agonist lixisenatide on postprandial glucose disposition was examined in conscious dogs to identify mechanisms for its improvement of meal tolerance in humans and examine the tissue disposition of meal-derived carbohydrate. Catheterization for measurement of hepatic balance occurred ≈16 days before study. After being fasted overnight, dogs received a subcutaneous injection of 1.5 µg/kg lixisenatide or vehicle (saline, control; n = 6/group). Thirty minutes later, they received an oral meal feeding (93.4 kJ; 19% protein, 71% glucose polymers, and 10% lipid). Acetaminophen was included in the meal in four control and five lixisenatide dogs for assessment of gastric emptying. Observations continued for 510 min; absorption was incomplete in lixisenatide at that point. The plasma acetaminophen area under the curve (AUC) in lixisenatide was 65% of that in control (P < 0.05). Absorption of the meal began within 15 min in control but was delayed until ≈30-45 min in lixisenatide. Lixisenatide reduced (P < 0.05) the postprandial arterial glucose AUC ≈54% and insulin AUC ≈44%. Net hepatic glucose uptake did not differ significantly between groups. Nonhepatic glucose uptake tended to be reduced by lixisenatide (6,151 ± 4,321 and 10,541 ± 1,854 µmol·kg(-1)·510 min(-1) in lixisenatide and control, respectively; P = 0.09), but adjusted (for glucose and insulin concentrations) values did not differ (18.9 ± 3.8 and 19.6 ± 7.9 l·kg(-1)·pmol(-1)·l(-1), lixisenatide and control, respectively; P = 0.94). Thus, lixisenatide delays gastric emptying, allowing more efficient disposal of the carbohydrate in the feeding without increasing liver glucose disposal. Lixisenatide could prove to be a valuable adjunct in treatment of postprandial hyperglycemia in impaired glucose tolerance or type 2 diabetes.


Sujet(s)
Glucose/métabolisme , Hypoglycémiants/pharmacologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Peptides/pharmacologie , Période post-prandiale/effets des médicaments et des substances chimiques , Acétaminophène/administration et posologie , Animaux , Conscience , Chiens , Femelle , Vidange gastrique/effets des médicaments et des substances chimiques , Glucagon/sang , Récepteur du peptide-1 similaire au glucagon , Insuline/sang , Mâle , Récepteurs au glucagon/agonistes
17.
Diabetes ; 62(10): 3553-60, 2013 Oct.
Article de Anglais | MEDLINE | ID: mdl-23835331

RÉSUMÉ

Endogenous hyperinsulinemia and insulin receptor (IR)/IGF-I receptor (IGF-IR) phosphorylation in tumors are associated with a worse prognosis in women with breast cancer. In vitro, insulin stimulation of the IR increases proliferation of breast cancer cells. However, in vivo studies demonstrating that IR activation increases tumor growth, independently of IGF-IR activation, are lacking. We hypothesized that endogenous hyperinsulinemia increases mammary tumor growth by directly activating the IR rather than the IGF-IR or hybrid receptors. We aimed to determine whether stimulating the IR with the insulin analog AspB10 could increase tumor growth independently of IGF-IR signaling. We induced orthotopic mammary tumors in control FVB/n and hyperinsulinemic MKR mice, and treated them with the insulin analog AspB10, recombinant human IGF-I, or vehicle. Tumors from mice with endogenous hyperinsulinemia were larger and had greater IR phosphorylation, but not IGF-IR phosphorylation, than those from control mice. Chronic AspB10 administration also increased tumor growth and IR (but not IGF-IR) phosphorylation in tumors. IGF-I led to activation of both the IGF-IR and IR and probably hybrid receptors. Our results demonstrate that IR phosphorylation increases tumor growth, independently of IGF-IR/hybrid receptor phosphorylation, and warrant consideration when developing therapeutics targeting the IGF-IR, but not the IR.


Sujet(s)
Tumeurs du sein/métabolisme , Diabète expérimental/traitement médicamenteux , Hyperinsulinisme/complications , Hypoglycémiants/effets indésirables , Facteur de croissance IGF-I/effets des médicaments et des substances chimiques , Insuline/analogues et dérivés , Syndrome métabolique X/complications , Thérapie moléculaire ciblée , Obésité/complications , Récepteur IGF de type 1/effets des médicaments et des substances chimiques , Animaux , Tumeurs du sein/étiologie , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Femelle , Humains , Hypoglycémiants/administration et posologie , Insuline/administration et posologie , Insuline/effets indésirables , Tumeurs expérimentales de la mamelle , Souris , Souris transgéniques , Thérapie moléculaire ciblée/tendances , Phosphorylation , Pronostic , Transduction du signal , Cellules cancéreuses en culture
18.
J Transl Med ; 11: 84, 2013 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-23537041

RÉSUMÉ

BACKGROUND: Lixisenatide is a glucagon-like peptide-1 analog which stimulates insulin secretion and inhibits glucagon secretion and gastric emptying. We investigated cardioprotective effects of lixisenatide in rodent models reflecting the clinical situation. METHODS: The acute cardiac effects of lixisenatide were investigated in isolated rat hearts subjected to brief ischemia and reperfusion. Effects of chronic treatment with lixisenatide on cardiac function were assessed in a modified rat heart failure model after only transient coronary occlusion followed by long-term reperfusion. Freshly isolated cardiomyocytes were used to investigate cell-type specific mechanisms of lixisenatide action. RESULTS: In the acute setting of ischemia-reperfusion, lixisenatide reduced the infarct-size/area at risk by 36% ratio without changes on coronary flow, left-ventricular pressure and heart rate. Treatment with lixisenatide for 10 weeks, starting after cardiac ischemia and reperfusion, improved left ventricular end-diastolic pressure and relaxation time and prevented lung congestion in comparison to placebo. No anti-fibrotic effect was observed. Gene expression analysis revealed a change in remodeling genes comparable to the ACE inhibitor ramipril. In isolated cardiomyocytes lixisenatide reduced apoptosis and increased fractional shortening. Glucagon-like peptide-1 receptor (GLP1R) mRNA expression could not be detected in rat heart samples or isolated cardiomyocytes. Surprisingly, cardiomyocytes isolated from GLP-1 receptor knockout mice still responded to lixisenatide. CONCLUSIONS: In rodent models, lixisenatide reduced in an acute setting infarct-size and improved cardiac function when administered long-term after ischemia-reperfusion injury. GLP-1 receptor independent mechanisms contribute to the described cardioprotective effect of lixisenatide. Based in part on these preclinical findings patients with cardiac dysfunction are currently being recruited for a randomized, double-blind, placebo-controlled, multicenter study with lixisenatide. TRIAL REGISTRATION: (ELIXA, ClinicalTrials.gov Identifier: NCT01147250).


Sujet(s)
Cardiotoniques/pharmacologie , Lésion de reperfusion myocardique/métabolisme , Peptides/pharmacologie , Androstadiènes/pharmacologie , Animaux , Modèles animaux de maladie humaine , Récepteur du peptide-1 similaire au glucagon , Défaillance cardiaque/traitement médicamenteux , Mâle , Souris , Souris knockout , Contraction myocardique/effets des médicaments et des substances chimiques , Myocarde/anatomopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley , Rat Wistar , Récepteurs au glucagon/métabolisme , Lésion d'ischémie-reperfusion/métabolisme , Transduction du signal , Wortmannine
19.
Arch Physiol Biochem ; 119(1): 1-14, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23373726

RÉSUMÉ

CONTEXT: Retrospective, observational studies have reported an association between diabetes treatment with insulin and a higher incidence of cancer. OBJECTIVE: Overview the literature for in vitro and in vivo studies of the metabolic and mitogenic properties of basal insulin analogues and assess the implications for clinical use. METHODS: Relevant studies were identified through PubMed and congress abstract database searches; data on metabolic and mitogenic signalling in relation to insulin treatment of diabetes are included in this review. RESULTS: The balance of evidence shows that although some analogues have demonstrated mitogenic potency in some in vitro studies in cancer cell lines, these findings do not translate to the in vivo setting in animals or to the clinical setting in humans. CONCLUSIONS: The current consensus is that there is no clinical or in vivo evidence to indicate that any commercially available insulin analogue has carcinogenic effects. Large-scale, prospective clinical and observational studies will further establish any potential link.


Sujet(s)
Complications du diabète/induit chimiquement , Diabète/traitement médicamenteux , Hypoglycémiants/effets indésirables , Insuline à longue durée d'action/pharmacologie , Tumeurs/induit chimiquement , Prolifération cellulaire , Bases de données factuelles , Complications du diabète/physiopathologie , Diabète/physiopathologie , Humains , Hypoglycémiants/administration et posologie , Hypoglycémiants/pharmacologie , Insuline glargine , Insuline à longue durée d'action/administration et posologie , Insuline à longue durée d'action/effets indésirables , Mitogènes/analyse , Phosphorylation , Liaison aux protéines , Récepteur IGF de type 1/métabolisme , Récepteur à l'insuline/métabolisme , Transduction du signal
20.
Regul Pept ; 164(2-3): 58-64, 2010 Sep 24.
Article de Anglais | MEDLINE | ID: mdl-20570597

RÉSUMÉ

The glucagon-like peptide-1 (GLP-1) receptor represents an established therapeutic target in type 2 diabetes mellitus (T2DM). Agents that activate this receptor improve glucose tolerance alongside a low risk of hypoglycaemia, and have the potential to modify disease progression. Lixisenatide is a new potent and selective GLP-1 receptor agonist currently in development. The preclinical pharmacological profile of Lixisenatide suggests actions that are highly relevant to the long-term maintenance of glucose homeostasis. Lixisenatide protected Ins-1 cells (a rat-derived beta-cell line) from both lipid- and cytokine-induced apoptosis. More importantly, Lixisenatide also prevented lipotoxicity-induced insulin depletion in human islets and preserved insulin production, storage and pancreatic beta-cell function in vitro. Enhancement of insulin biosynthesis and pancreatic beta-cell volume could also be demonstrated in animal models of type 2 diabetes. The improvement of glucose-stimulated insulin secretion provided by Lixisenatide occurred in a strictly glucose-dependent manner. In animal models of diabetes, Lixisenatide improved basal blood glucose and HbA(1c) with a rapid onset and sustained duration of action, and prevented the deterioration of pancreatic responsiveness and glucose homeostasis. Lixisenatide also delayed gastric emptying and reduced food intake. The efficacy/safety profile of Lixisenatide is currently being studied further in an extensive ongoing Phase III clinical study programme. This article reviews the preclinical pharmacological profile of Lixisenatide.


Sujet(s)
Diabète de type 2/traitement médicamenteux , Hypoglycémiants/usage thérapeutique , Peptides/usage thérapeutique , Récepteurs au glucagon/antagonistes et inhibiteurs , Animaux , Récepteur du peptide-1 similaire au glucagon , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE