Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 64(13): 8942-8950, 2021 07 08.
Article in English | MEDLINE | ID: mdl-33944562

ABSTRACT

Here, we describe the molecular engineering of insulin icodec to achieve a plasma half-life of 196 h in humans, suitable for once-weekly subcutaneously administration. Insulin icodec is based on re-engineering of the ultra-long oral basal insulin OI338 with a plasma half-life of 70 h in humans. This systematic re-engineering was accomplished by (1) further increasing the albumin binding by changing the fatty diacid from a 1,18-octadecanedioic acid (C18) to a 1,20-icosanedioic acid (C20) and (2) further reducing the insulin receptor affinity by the B16Tyr → His substitution. Insulin icodec was selected by screening for long intravenous plasma half-life in dogs while ensuring glucose-lowering potency following subcutaneous administration in rats. The ensuing structure-activity relationship resulted in insulin icodec. In phase-2 clinical trial, once-weekly insulin icodec provided safe and efficacious glycemic control comparable to once-daily insulin glargine in type 2 diabetes patients. The structure-activity relationship study leading to insulin icodec is presented here.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Animals , Dogs , Drug Administration Schedule , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/chemistry , Injections, Intravenous , Injections, Subcutaneous , Insulin/administration & dosage , Insulin/analogs & derivatives , Male , Rats , Rats, Sprague-Dawley
2.
J Med Chem ; 64(1): 616-628, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33356257

ABSTRACT

Recently, the first basal oral insulin (OI338) was shown to provide similar treatment outcomes to insulin glargine in a phase 2a clinical trial. Here, we report the engineering of a novel class of basal oral insulin analogues of which OI338, 10, in this publication, was successfully tested in the phase 2a clinical trial. We found that the introduction of two insulin substitutions, A14E and B25H, was needed to provide increased stability toward proteolysis. Ultralong pharmacokinetic profiles were obtained by attaching an albumin-binding side chain derived from octadecanedioic (C18) or icosanedioic acid (C20) to the lysine in position B29. Crucial for obtaining the ultralong PK profile was also a significant reduction of insulin receptor affinity. Oral bioavailability in dogs indicated that C18-based analogues were superior to C20-based analogues. These studies led to the identification of the two clinical candidates OI338 and OI320 (10 and 24, respectively).


Subject(s)
Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Acylation , Administration, Oral , Amino Acid Sequence , Animals , Biological Availability , Delayed-Action Preparations , Dogs , Half-Life , Humans , Hypoglycemic Agents/pharmacokinetics , Insulin/chemistry , Insulin/pharmacokinetics , Rats
3.
Nat Commun ; 11(1): 3746, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32719315

ABSTRACT

Recently, the clinical proof of concept for the first ultra-long oral insulin was reported, showing efficacy and safety similar to subcutaneously administered insulin glargine. Here, we report the molecular engineering as well as biological and pharmacological properties of these insulin analogues. Molecules were designed to have ultra-long pharmacokinetic profile to minimize variability in plasma exposure. Elimination plasma half-life of ~20 h in dogs and ~70 h in man is achieved by a strong albumin binding, and by lowering the insulin receptor affinity 500-fold to slow down receptor mediated clearance. These insulin analogues still stimulate efficient glucose disposal in rats, pigs and dogs during constant intravenous infusion and euglycemic clamp conditions. The albumin binding facilitates initial high plasma exposure with a concomitant delay in distribution to peripheral tissues. This slow appearance in the periphery mediates an early transient hepato-centric insulin action and blunts hypoglycaemia in dogs in response to overdosing.


Subject(s)
Insulin/administration & dosage , Protein Engineering , Administration, Oral , Amino Acid Sequence , Animals , Blood Glucose/metabolism , Computer Simulation , Dogs , Dose-Response Relationship, Drug , Drug Overdose/blood , Glucose Clamp Technique , Half-Life , Humans , Hyperinsulinism/drug therapy , Hypoglycemia/diagnosis , Insulin/analogs & derivatives , Insulin/chemistry , Insulin/pharmacokinetics , Male , Protein Stability , Proteolysis , Rats, Sprague-Dawley , Swine , Treatment Outcome
5.
Pharm Res ; 36(3): 49, 2019 Feb 11.
Article in English | MEDLINE | ID: mdl-30746556

ABSTRACT

PURPOSE: Fast-acting insulin aspart (faster aspart) is a novel formulation of insulin aspart containing two additional excipients: niacinamide, to increase early absorption, and L-arginine, to optimize stability. The aim of this study was to evaluate the impact of niacinamide on insulin aspart absorption and to investigate the mechanism of action underlying the accelerated absorption. METHODS: The impact of niacinamide was assessed in pharmacokinetic analyses in pigs and humans, small angle X-ray scattering experiments, trans-endothelial transport assays, vascular tension measurements, and subcutaneous blood flow imaging. RESULTS: Niacinamide increased the rate of early insulin aspart absorption in pigs, and pharmacokinetic modelling revealed this effect to be most pronounced up to ~30-40 min after injection in humans. Niacinamide increased the relative monomer fraction of insulin aspart by ~35%, and the apparent permeability of insulin aspart across an endothelial cell barrier by ~27%. Niacinamide also induced a concentration-dependent vasorelaxation of porcine arteries, and increased skin perfusion in pigs. CONCLUSION: Niacinamide mediates the acceleration of initial insulin aspart absorption, and the mechanism of action appears to be multifaceted. Niacinamide increases the initial abundance of insulin aspart monomers and transport of insulin aspart after subcutaneous administration, and also mediates a transient, local vasodilatory effect.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/pharmacokinetics , Insulin Aspart/pharmacokinetics , Niacinamide/pharmacology , Subcutaneous Absorption/drug effects , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/blood , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Female , Humans , Hypoglycemic Agents/administration & dosage , Injections, Subcutaneous , Insulin Aspart/administration & dosage , Models, Biological , Regional Blood Flow/drug effects , Scattering, Small Angle , Subcutaneous Tissue/blood supply , Subcutaneous Tissue/drug effects , Subcutaneous Tissue/metabolism , Sus scrofa , Vasodilation/drug effects , X-Ray Diffraction
6.
Protein Sci ; 22(3): 296-305, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23281053

ABSTRACT

Insulin is a key hormone controlling glucose homeostasis. All known vertebrate insulin analogs have a classical structure with three 100% conserved disulfide bonds that are essential for structural stability and thus the function of insulin. It might be hypothesized that an additional disulfide bond may enhance insulin structural stability which would be highly desirable in a pharmaceutical use. To address this hypothesis, we designed insulin with an additional interchain disulfide bond in positions A10/B4 based on Cα-Cα distances, solvent exposure, and side-chain orientation in human insulin (HI) structure. This insulin analog had increased affinity for the insulin receptor and apparently augmented glucodynamic potency in a normal rat model compared with HI. Addition of the disulfide bond also resulted in a 34.6°C increase in melting temperature and prevented insulin fibril formation under high physical stress even though the C-terminus of the B-chain thought to be directly involved in fibril formation was not modified. Importantly, this analog was capable of forming hexamer upon Zn addition as typical for wild-type insulin and its crystal structure showed only minor deviations from the classical insulin structure. Furthermore, the additional disulfide bond prevented this insulin analog from adopting the R-state conformation and thus showing that the R-state conformation is not a prerequisite for binding to insulin receptor as previously suggested. In summary, this is the first example of an insulin analog featuring a fourth disulfide bond with increased structural stability and retained function.


Subject(s)
Antigens, CD/metabolism , Cystine/chemistry , Glucose/metabolism , Hypoglycemic Agents/chemistry , Insulin, Regular, Human/analogs & derivatives , Receptor, Insulin/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Amino Acid Substitution , Animals , Biological Transport/drug effects , Blood Glucose/analysis , Cells, Cultured , Cystine/metabolism , Dose-Response Relationship, Drug , Drug Stability , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin, Regular, Human/chemistry , Insulin, Regular, Human/genetics , Insulin, Regular, Human/metabolism , Insulin, Regular, Human/pharmacology , Mutant Proteins/administration & dosage , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutant Proteins/pharmacology , Protein Conformation , Protein Stability , Rats , Rats, Mutant Strains , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Zinc/metabolism
7.
Nature ; 437(7061): 975-80, 2005 Oct 13.
Article in English | MEDLINE | ID: mdl-16222292

ABSTRACT

Animals and higher plants express endogenous peptide antibiotics called defensins. These small cysteine-rich peptides are active against bacteria, fungi and viruses. Here we describe plectasin-the first defensin to be isolated from a fungus, the saprophytic ascomycete Pseudoplectania nigrella. Plectasin has primary, secondary and tertiary structures that closely resemble those of defensins found in spiders, scorpions, dragonflies and mussels. Recombinant plectasin was produced at a very high, and commercially viable, yield and purity. In vitro, the recombinant peptide was especially active against Streptococcus pneumoniae, including strains resistant to conventional antibiotics. Plectasin showed extremely low toxicity in mice, and cured them of experimental peritonitis and pneumonia caused by S. pneumoniae as efficaciously as vancomycin and penicillin. These findings identify fungi as a novel source of antimicrobial defensins, and show the therapeutic potential of plectasin. They also suggest that the defensins of insects, molluscs and fungi arose from a common ancestral gene.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Fungi/chemistry , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Cloning, Molecular , DNA, Complementary/genetics , Defensins/chemistry , Disease Models, Animal , Fungi/genetics , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/physiology , Humans , Mice , Molecular Sequence Data , Peptides , Pneumococcal Infections/drug therapy , Pneumococcal Infections/microbiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
8.
J Biol Chem ; 277(21): 18245-8, 2002 May 24.
Article in English | MEDLINE | ID: mdl-11923277

ABSTRACT

Adaptation to efficient heterologous expression is a prerequisite for recombinant proteins to fulfill their clinical and biotechnological potential. We describe a rational strategy to optimize the secretion efficiency in yeast of an insulin precursor by structure-based engineering of the folding stability. The yield of a fast-acting insulin analogue (Asp(B28)) expressed in yeast was enhanced 5-fold by engineering a specific interaction between an aromatic amino acid in the connecting peptide and a phenol binding site in the hydrophobic core of the molecule. This insulin precursor is characterized by significantly enhanced folding stability. The improved folding properties enhanced the secretion efficiency of the insulin precursor from 10 to 50%. The precursor remains fully in vitro convertible to mature fast-acting insulin.


Subject(s)
Insulin/genetics , Protein Engineering , Saccharomyces cerevisiae/genetics , Insulin/chemistry , Models, Molecular , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...