1
|
Gare CL, White AM, Malins LR. From lead to market: chemical approaches to transform peptides into therapeutics. Trends Biochem Sci 2025; 50:467-480. [PMID: 40011178 DOI: 10.1016/j.tibs.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/28/2025]
Abstract
Peptides are a powerful drug modality with potential to access difficult targets. This recognition underlies their growth in the global pharmaceutical market, with peptides representing ~8% of drugs approved by the FDA over the past decade. Currently, the peptide therapeutic landscape is evolving, with high-throughput display technologies driving the identification of peptide leads with enhanced diversity. Yet, chemical modifications remain essential for improving the 'drug-like' properties of peptides and ultimately translating leads to market. In this review, we explore two recent therapeutic candidates (semaglutide, a peptide hormone analogue, and MK-0616, an mRNA display-derived candidate) as case studies that highlight general approaches to improving pharmacokinetics (PK) and potency. We also emphasize the critical link between advances in medicinal chemistry and the optimisation of highly efficacious peptide therapeutics.
Collapse
Affiliation(s)
- Caitlin L Gare
- Research School of Chemistry, Australian National University, Canberra 2601, Australian Capital Territory, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra 2601, Australian Capital Territory, Australia
| | - Andrew M White
- Research School of Chemistry, Australian National University, Canberra 2601, Australian Capital Territory, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra 2601, Australian Capital Territory, Australia
| | - Lara R Malins
- Research School of Chemistry, Australian National University, Canberra 2601, Australian Capital Territory, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra 2601, Australian Capital Territory, Australia.
| |
Collapse
|
2
|
AL-Noshokaty TM, Abdelhamid R, Abdelmaksoud NM, Khaled A, Hossam M, Ahmed R, Saber T, Khaled S, Elshaer SS, Abulsoud AI. Unlocking the multifaceted roles of GLP-1: Physiological functions and therapeutic potential. Toxicol Rep 2025; 14:101895. [PMID: 39911322 PMCID: PMC11795145 DOI: 10.1016/j.toxrep.2025.101895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025] Open
Abstract
Glucagon (GCG) like peptide 1 (GLP-1) has emerged as a powerful player in regulating metabolism and a promising therapeutic target for various chronic diseases. This review delves into the physiological roles of GLP-1, exploring its impact on glucose homeostasis, insulin secretion, and satiety. We examine the compelling evidence supporting GLP-1 receptor agonists (GLP-1RAs) in managing type 2 diabetes (T2D), obesity, and other diseases. The intricate molecular mechanisms underlying GLP-1RAs are explored, including their interactions with pathways like extracellular signal-regulated kinase 1/2 (ERK1/2), activated protein kinase (AMPK), cyclic adenine monophosphate (cAMP), mitogen-activated protein kinase (MAPK), and protein kinase C (PKC). Expanding our understanding, the review investigates the potential role of GLP-1 in cancers. Also, microribonucleic acid (RNA) (miRNAs), critical regulators of gene expression, are introduced as potential modulators of GLP-1 signaling. We delve into the link between miRNAs and T2D obesity and explore specific miRNA examples influencing GLP-1R function. Finally, the review explores the rationale for seeking alternatives to GLP-1RAs and highlights natural products with promising GLP-1 modulatory effects.
Collapse
Affiliation(s)
- Tohada M. AL-Noshokaty
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Rehab Abdelhamid
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | | | - Aya Khaled
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Mariam Hossam
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Razan Ahmed
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Toka Saber
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Shahd Khaled
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Shereen Saeid Elshaer
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Ahmed I. Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt
- Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| |
Collapse
|
3
|
Tordrup EK, Gadgaard S, Windeløv J, Holst JJ, Gasbjerg LS, Hartmann B, Rosenkilde MM. Development of a long-acting unbiased GIP receptor agonist for studies of GIP's role in bone metabolism. Biochem Pharmacol 2025; 236:116893. [PMID: 40132763 DOI: 10.1016/j.bcp.2025.116893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/26/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
The incretin hormone glucose-dependent insulinotropic polypeptide (GIP) stimulates bone remodeling postprandially. Species variations complicate the development of long-acting agonists with similar effects on rodent and human GIP receptors (GIPR). We created a series of long-acting molecules suitable for rat studies based on human GIP, stabilized with Aib insertion in position 2, lipidations in the middle region (compounds 1-4: positions 14/16/17/20) or the C-terminus (compound 5: position 40), and elongation with an exendin-4 tail in the C-terminus (Cex). The compounds were tested in vitro on the human and rat GIPR for cAMP accumulation, beta-arrestin recruitment and internalization. Pharmacokinetic profiling in rats was completed for two compounds, and one was selected for bone remodeling studies in rats (measurements of C-terminal telopeptide (CTX) and procollagen type 1 N-propeptide). All five compounds retained the potency and efficacy of native (human and rat) GIP in cAMP accumulation and arrestin recruitment on human and rat GIPR with no differences in relative activities from native GIP. Only compound 3 induced internalization like species-matched GIP on respective receptors and was chosen for in vivo assessments in rats. Mean T1/2 was 9.1 h, and it decreased plasma levels of CTX compared to vehicle treatment following 1000 µg·kg-1 injections. In conclusion, the long-acting, unbiased compound 3 (hGIP(1-30-Cex)/Aib2/C16-diacid moiety in position 17), with retained activity for the human and rat GIPR, is suitable for bone remodeling studies in rats; hence, a useful tool compound for future research of GIP's therapeutic potential in bone-related diseases.
Collapse
Affiliation(s)
- Esther Karen Tordrup
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | | | - Johanne Windeløv
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Bainan Biotech ApS, Copenhagen, Denmark.
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Lærke Smidt Gasbjerg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Mette Marie Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
4
|
Gupta S, Gupta A, Mukherjee M, Bose S, Sinha S. Chemical Insights into Oligonucleotide-Protein Binding for Therapeutic Applications. J Med Chem 2025; 68:9848-9863. [PMID: 40332202 DOI: 10.1021/acs.jmedchem.5c00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Plasma protein binding is an important determinant in the clinical success of oligonucleotide-based drugs. Optimal protein binding of the oligonucleotide is critical to its tissue distribution and retention by preventing renal excretion. This property can be modulated through suitable chemical modifications depending on the oligonucleotide backbone to achieve a balanced pharmacokinetic profile and minimize off-target effects. The macromolecular structure of the oligonucleotide leads to dynamic protein binding characteristics as compared to small-molecule-based drugs, which are not associated with additional barriers such as intracellular delivery. This perspective provides insight into the diverse plasma protein interactions of various classes of oligonucleotides and explores chemical strategies for modulating these interactions. Furthermore, we have discussed different methods for the quantification of plasma protein binding along with the correlation of chemistry and therapeutic outcomes of FDA-approved oligonucleotides.
Collapse
Affiliation(s)
- Shalini Gupta
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Abhishek Gupta
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Maria Mukherjee
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Sritama Bose
- Medical Research Council, Nucleic Acid Therapy Accelerator, (UKRI) Research Complex at Harwell (RCaH), Rutherford Appleton Laboratory, Harwell OX11 0FA, U.K
| | - Surajit Sinha
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| |
Collapse
|
5
|
Roberts TD, Hutchinson DS, Wootten D, De Blasio MJ, Ritchie RH. Advances in incretin therapies for targeting cardiovascular disease in diabetes. J Mol Cell Cardiol 2025; 202:102-115. [PMID: 40086589 DOI: 10.1016/j.yjmcc.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
The global prevalence of obesity is skyrocketing at an alarming rate, with recent data estimating that one-in-eight people are now living with the disease. Obesity is a chronic metabolic disorder that shares underlying pathophysiology with other metabolically-linked diseases such as type 2 diabetes mellitus, cardiovascular disease and diabetic cardiomyopathy. There is a distinct correlation between type 2 diabetes status and the likelihood of heart failure. Of note, there is an apparent sexual dimorphism, with women disproportionately affected with respect to the degree of severity of the cardiac phenotype of diabetic cardiomyopathy that results from diabetes. The current pharmacotherapies available for the attenuation of hyperglycaemia in type 2 diabetes are not always effective, and have varying degrees of efficacy in the setting of heart failure. Insulin can worsen heart failure prognosis whereas metformin, sodium-glucose cotransporter 2 inhibitors (SGLT2i) and more recently, glucagon-like peptide-1 receptor agonists (GLP-1RAs), have demonstrated cardioprotection with their administration. This review will highlight the advancement of incretin therapies for individuals with diabetes and heart failure and explore newly-reported evidence of the clinical usefulness of GLP-1R agonists in this distinct phenotype of heart failure.
Collapse
Affiliation(s)
- Timothy D Roberts
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Dana S Hutchinson
- Metabolic G Protein-Coupled Receptor Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Denise Wootten
- Metabolic G Protein-Coupled Receptor Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Miles J De Blasio
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| | - Rebecca H Ritchie
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| |
Collapse
|
6
|
Jing W, Peng L, Song S, Liu J, Tai W. A New Protractor Potentiates Glucagon-Like Peptide 1 with Slow-Release Depot and Long-Term Action. J Med Chem 2025; 68:7341-7352. [PMID: 40118774 DOI: 10.1021/acs.jmedchem.4c02970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Bioactive peptides display a number of favorable features as therapeutics, but their usage is challenging due to the low metabolic stability and rapid renal clearance. The small-molecule protractor, which functions by the noncovalent binding with serum albumin and protection against systemic clearance, is an attractive tool to elongate peptides' half-life. Herein, we investigated coomassie brilliant blue (CBB) as a new protractor for the half-life extension of clinically relevant glucagon-like peptide 1 (GLP-1). A series of GLP-1 analogues differentiating with CBB linkers and acylation positions are described. One particularly interesting analogue (coomatide 13) exhibits sub-picomolar potency in vitro and long-term control of glucose homeostasis in mice. A protraction mechanism study reveals that CBB has a high affinity to albumin and pan-interaction with other matrix proteins, enabling to protract peptides in both systemic circulation and the subcutaneous depot. Our study demonstrates that the specific affinity to albumin is not a prerequisite for peptide protraction, and pan-binders might be advantageous.
Collapse
Affiliation(s)
- Weina Jing
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Lei Peng
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Shiwei Song
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Jiaqi Liu
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Wanyi Tai
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
7
|
Yu M, Zhang C, Xu H, Dong Y, Zhu H, Xia C, Feng J. Design of a novel long-acting insulin analogs by acetylation modification and compared with insulin Icodec. Sci Rep 2025; 15:9408. [PMID: 40108309 PMCID: PMC11923205 DOI: 10.1038/s41598-025-94014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Insulin is a potent medication for managing diabetes, yet its short half-life requires daily administration. Currently, Novo Nordisk's icodec is the sole insulin available on the market that requires administration only once a week. Insulin icodec, developed by Novo Nordisk through amino acid mutations and fatty acid side chain modifications, has demonstrated the capability to control blood glucose levels on a once-weekly basis. To improve its efficacy, we modified the acylation side chain of icodec to generate insulin analogs appropriate for weekly dosing. A promising insulin analog, TBE001-A-S033, was synthesized and conjugated, and its efficacy was assessed in ICR and db/db mice. TBE001-A-S033 prolonged blood glucose control in ICR mice and exhibited a comparable blood glucose trend to insulin icodec in db/db mice. These findings suggest that TBE001-A-S033 possesses a favorable hypoglycemic effect and a differential half-life across species compared to insulin icodec, indicating its potential for once-weekly use in humans. This preclinical investigation indicates that TBE001-A-S033 may serve as an effective therapeutic for type 2 diabetes mellitus (T2DM).
Collapse
Affiliation(s)
- Min Yu
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Lianyungang, Jiangsu, People's Republic of China
| | - Chuanzhi Zhang
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Lianyungang, Jiangsu, People's Republic of China
| | - Hongjiang Xu
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Lianyungang, Jiangsu, People's Republic of China
| | - Yuanzhen Dong
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
- Shanghai Duomirui Biotechnology Ltd, No.285 Gebaini Road, Pudong New Area, Shanghai, 201203, China
| | - Hongxiang Zhu
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Chunguang Xia
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Lianyungang, Jiangsu, People's Republic of China
| | - Jun Feng
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China.
- Shanghai Duomirui Biotechnology Ltd, No.285 Gebaini Road, Pudong New Area, Shanghai, 201203, China.
| |
Collapse
|
8
|
Duran M, Willis JR, Dalvi N, Fokakis Z, Virkus SA, Hardaway JA. Integration of Glucagon-Like Peptide 1 Receptor Actions Through the Central Amygdala. Endocrinology 2025; 166:bqaf019. [PMID: 39888375 PMCID: PMC11850305 DOI: 10.1210/endocr/bqaf019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/31/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025]
Abstract
Understanding the detailed mechanism of action of glucagon-like peptide 1 receptor (GLP-1R) agonists on distinct topographic and genetically defined brain circuits is critical for improving the efficacy and mitigating adverse side effects of these compounds. In this mini-review, we propose that the central nucleus of the amygdala (CeA) is a critical mediator of GLP-1R agonist-driven hypophagia. Here, we review the extant literature demonstrating CeA activation via GLP-1R agonists across multiple species and through multiple routes of administration. The precise role of GLP-1Rs within the CeA is unclear but the site-specific GLP-1Rs may mediate distinct behavioral and physiological hallmarks of GLP-1R agonists on food intake. Thus, we propose important novel directions and methods to test the role of the CeA in mediating GLP-1R actions.
Collapse
Affiliation(s)
- Miguel Duran
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer R Willis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nilay Dalvi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zoe Fokakis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sonja A Virkus
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - J Andrew Hardaway
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
9
|
Kristensen JB, Elster L, Lundh M, Ballarín-González B, Alexopoulou F, Kræmer M, Jensen DM, Leurs U, Nielsen JC, Hansen HH, Haanes KA, Degn M. Pipeline for development of acylated peptide based CGRP receptor antagonist with extended half-life for migraine treatment. Sci Rep 2025; 15:1870. [PMID: 39805895 PMCID: PMC11730311 DOI: 10.1038/s41598-024-84547-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
Migraine is a debilitating headache disorder. The disease has neurovascular origin and migraine attacks can be elicited by vasodilative neuropeptides such as alpha calcitonin gene-related peptide (αCGRP). Antagonizing CGRP actions in migraine patients has proven clinically efficient. Here, we present a pipeline for development of a peptide-based hCGRP receptor antagonist with increased half-life capable of antagonising the vasodilatory effect of hαCGRP. A series of hαCGRP8-37 analogues carrying a C18-or C20-diacid lipidation was screened for their antagonism against the hCGRP receptor. hαCGRP8-37 analogues with a C20-diacid were 2-6 fold more potent than analogues conjugated with a C18-diacid. Half-life of hαCGRP8-37 analogues carrying a C20-diacid was estimated in mice in a pilot study (n = 1-2). Half-lives ranged from 7.3 to 13.7 h. An hαCGRP8-37 analogue conjugated with a C20 diacid at position 25 was subjected to an amino acid substitution scan to identify mutations that could further enhance hCGRP receptor antagonism. Substituting alanine with serine at position 36 resulted in a ~ 4 fold gain of potency. Vasodilative actions of hαCGRP were successfully antagonized by hαCGRP8-37 analogues carrying a C20 diacid at position 25. Our findings demonstrate that lipidation can improve hαCGRP8-37 pharmacokinetics while maintaining hαCGRP antagonism, thus demonstrating potential for a peptide-based migraine treatment strategy.
Collapse
Affiliation(s)
- Jens Bjelke Kristensen
- Gubra ApS, Hørsholm Kongevej 11B, DK-2970, Hørsholm, Denmark.
- Sensory Biology Unit, Translational Research Centre, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark.
| | - Lisbeth Elster
- Gubra ApS, Hørsholm Kongevej 11B, DK-2970, Hørsholm, Denmark
| | - Morten Lundh
- Gubra ApS, Hørsholm Kongevej 11B, DK-2970, Hørsholm, Denmark
| | | | - Flora Alexopoulou
- Gubra ApS, Hørsholm Kongevej 11B, DK-2970, Hørsholm, Denmark
- Novo Nordisk, Måløv, Denmark
| | - Martin Kræmer
- Gubra ApS, Hørsholm Kongevej 11B, DK-2970, Hørsholm, Denmark
| | | | - Ulrike Leurs
- Gubra ApS, Hørsholm Kongevej 11B, DK-2970, Hørsholm, Denmark
| | | | - Henrik H Hansen
- Gubra ApS, Hørsholm Kongevej 11B, DK-2970, Hørsholm, Denmark
| | - Kristian A Haanes
- Sensory Biology Unit, Translational Research Centre, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Biology, Section of Cell Biology and Physiology, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Matilda Degn
- Gubra ApS, Hørsholm Kongevej 11B, DK-2970, Hørsholm, Denmark.
| |
Collapse
|
10
|
Qi S, Liu Z, Suyama K, Tsuchiya Y, Canarejo J, Phan KQ, Yutsudo N, Shimada A, Hirota T, Ieiri I, Kishimura A, Muraoka T, Nose T, Mori T, Katayama Y. Ligand Design with Accelerated Disulfide Formation with Serum Albumin to Extend Blood Retention. ACS Med Chem Lett 2025; 16:144-148. [PMID: 39811133 PMCID: PMC11726352 DOI: 10.1021/acsmedchemlett.4c00503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
We proposed a novel ligand for the interaction with human serum albumin (HSA) to extend the blood half-life of small molecular weight therapeutics. The ligand features an alkyl chain and an activated disulfide to allow binding to the hydrophobic pockets of HSA and the formation of disulfide to Cys34 of HSA, thereby minimizing the initial renal clearance. The dual nature of the ligand-HSA bonding was expected to give the ligand long blood retention. After 1 min of mixing with HSA, the ligand showed higher binding (1.7 times) than that of a control ligand (containing only activated disulfide). After intravenous injection to mice, the ligand half-lives were 1.6 and 9.2 times longer than those of control ligands with the active disulfide alone and with the alkyl chain alone, respectively. The proposed ligand has the potential to act as a platform for extending the half-life of small therapeutics in vivo.
Collapse
Affiliation(s)
- Song Qi
- Graduate
school of Systems Life Sciences, Kyushu
University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Zixuan Liu
- Graduate
school of Systems Life Sciences, Kyushu
University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Keitaro Suyama
- Faculty
of Arts and Science, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yuichi Tsuchiya
- Department
of Pharmacy, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jedidiah Canarejo
- Department
of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Khanh Quoc Phan
- Graduate
school of Systems Life Sciences, Kyushu
University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Noriko Yutsudo
- Advanced
Research Initiative, Research Promotion Unit, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Atsushi Shimada
- Advanced
Research Initiative, Research Promotion Unit, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Hirota
- Faculty
of Pharmaceutical Sciences Graduate School of Pharmaceutical Sciences, Kyushu university, 3-1-1 Maidashi, Higashi-ku,
Fukuoka 812-8582, Japan
| | - Ichiro Ieiri
- Faculty
of Pharmaceutical Sciences Graduate School of Pharmaceutical Sciences, Kyushu university, 3-1-1 Maidashi, Higashi-ku,
Fukuoka 812-8582, Japan
| | - Akihiro Kishimura
- Department
of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takahiro Muraoka
- Department
of Applied Chemistry, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan
| | - Takeru Nose
- Faculty
of Arts and Science, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takeshi Mori
- Department
of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yoshiki Katayama
- Department
of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
11
|
Zhu W, Tanday N, Lafferty RA, Flatt PR, Irwin N. Novel enzyme-resistant pancreatic polypeptide analogs evoke pancreatic beta-cell rest, enhance islet cell turnover, and inhibit food intake in mice. Biofactors 2024; 50:1101-1112. [PMID: 38635341 PMCID: PMC11627468 DOI: 10.1002/biof.2059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Pancreatic polypeptide (PP) is a postprandial hormone secreted from pancreatic islets that activates neuropeptide Y4 receptors (NPY4Rs). PP is known to induce satiety but effects at the level of the endocrine pancreas are less well characterized. In addition, rapid metabolism of PP by dipeptidyl peptidase-4 (DPP-4) limits the investigation of the effects of the native peptide. Therefore, in the present study, five novel amino acid substituted and/or fatty acid derivatized PP analogs were synthesized, namely [P3]PP, [K13Pal]PP, [P3,K13Pal]PP, [N-Pal]PP, and [N-Pal,P3]PP, and their impact on pancreatic beta-cell function, as well as appetite regulation and glucose homeostasis investigated. All PP analogs displayed increased resistance to DPP-4 degradation. In addition, all peptides inhibited alanine-induced insulin secretion from BRIN-BD11 beta cells. Native PP and related analogs (10-8 and 10-6 M), and especially [P3]PP and [K13Pal]PP, significantly protected against cytokine-induced beta-cell apoptosis and promoted cellular proliferation, with effects dependent on the NPY4R for all peptides barring [N-Pal,P3]PP. In mice, all peptides, except [N-Pal]PP and [N-Pal,P3]PP, evoked a dose-dependent (25, 75, and 200 nmol/kg) suppression of appetite, with native PP and [P3]PP further augmenting glucagon-like peptide-1 (GLP-1) and cholecystokinin (CCK) induced reductions of food intake. The PP peptides had no obvious detrimental effect on glucose tolerance and they did not noticeably impair the glucose-regulatory actions of GLP-1 or CCK. In conclusion, Pro3 amino acid substitution of PP, either alone or together with mid-chain acylation, creates PP analogs with benefits on beta-cell rest, islet cell turnover, and energy regulation that may be applicable to the treatment of diabetes and obesity.
Collapse
Affiliation(s)
- Wuyun Zhu
- Diabetes Research CentreSchools of Biomedical Sciences and Pharmacy & Pharmaceutical Sciences, Ulster UniversityColeraineUK
| | - Neil Tanday
- Diabetes Research CentreSchools of Biomedical Sciences and Pharmacy & Pharmaceutical Sciences, Ulster UniversityColeraineUK
| | - Ryan A. Lafferty
- Diabetes Research CentreSchools of Biomedical Sciences and Pharmacy & Pharmaceutical Sciences, Ulster UniversityColeraineUK
| | - Peter R. Flatt
- Diabetes Research CentreSchools of Biomedical Sciences and Pharmacy & Pharmaceutical Sciences, Ulster UniversityColeraineUK
| | - Nigel Irwin
- Diabetes Research CentreSchools of Biomedical Sciences and Pharmacy & Pharmaceutical Sciences, Ulster UniversityColeraineUK
| |
Collapse
|
12
|
Sass F, Ma T, Ekberg JH, Kirigiti M, Ureña MG, Dollet L, Brown JM, Basse AL, Yacawych WT, Burm HB, Andersen MK, Nielsen TS, Tomlinson AJ, Dmytiyeva O, Christensen DP, Bader L, Vo CT, Wang Y, Rausch DM, Kristensen CK, Gestal-Mato M, In Het Panhuis W, Sjøberg KA, Kernodle S, Petersen JE, Pavlovskyi A, Sandhu M, Moltke I, Jørgensen ME, Albrechtsen A, Grarup N, Babu MM, Rensen PCN, Kooijman S, Seeley RJ, Worthmann A, Heeren J, Pers TH, Hansen T, Gustafsson MBF, Tang-Christensen M, Kilpeläinen TO, Myers MG, Kievit P, Schwartz TW, Hansen JB, Gerhart-Hines Z. NK2R control of energy expenditure and feeding to treat metabolic diseases. Nature 2024; 635:987-1000. [PMID: 39537932 PMCID: PMC11602716 DOI: 10.1038/s41586-024-08207-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
The combination of decreasing food intake and increasing energy expenditure represents a powerful strategy for counteracting cardiometabolic diseases such as obesity and type 2 diabetes1. Yet current pharmacological approaches require conjugation of multiple receptor agonists to achieve both effects2-4, and so far, no safe energy-expending option has reached the clinic. Here we show that activation of neurokinin 2 receptor (NK2R) is sufficient to suppress appetite centrally and increase energy expenditure peripherally. We focused on NK2R after revealing its genetic links to obesity and glucose control. However, therapeutically exploiting NK2R signalling has previously been unattainable because its endogenous ligand, neurokinin A, is short-lived and lacks receptor specificity5,6. Therefore, we developed selective, long-acting NK2R agonists with potential for once-weekly administration in humans. In mice, these agonists elicit weight loss by inducing energy expenditure and non-aversive appetite suppression that circumvents canonical leptin signalling. Additionally, a hyperinsulinaemic-euglycaemic clamp reveals that NK2R agonism acutely enhances insulin sensitization. In diabetic, obese macaques, NK2R activation significantly decreases body weight, blood glucose, triglycerides and cholesterol, and ameliorates insulin resistance. These findings identify a single receptor target that leverages both energy-expending and appetite-suppressing programmes to improve energy homeostasis and reverse cardiometabolic dysfunction across species.
Collapse
Affiliation(s)
- Frederike Sass
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
| | - Tao Ma
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jeppe H Ekberg
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Embark Laboratories, Copenhagen, Denmark
| | - Melissa Kirigiti
- Division of Metabolic Health and Disease, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Mario G Ureña
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Lucile Dollet
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jenny M Brown
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Astrid L Basse
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Warren T Yacawych
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Hayley B Burm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Mette K Andersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Thomas S Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Oksana Dmytiyeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Dan P Christensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Embark Laboratories, Copenhagen, Denmark
| | - Lindsay Bader
- Division of Metabolic Health and Disease, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Camilla T Vo
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Neuroscience Academy Denmark, Copenhagen, Denmark
| | - Yaxu Wang
- Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
- Center of Excellence for Data Driven Discovery, Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Dylan M Rausch
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie K Kristensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - María Gestal-Mato
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Wietse In Het Panhuis
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory of Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Kim A Sjøberg
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Stace Kernodle
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jacob E Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Artem Pavlovskyi
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Manbir Sandhu
- Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
- Center of Excellence for Data Driven Discovery, Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ida Moltke
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Marit E Jørgensen
- Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Herlev, Denmark
- Centre for Public Health in Greenland, National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
- Steno Diabetes Center Greenland, Nuuk, Greenland
| | - Anders Albrechtsen
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - M Madan Babu
- Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
- Center of Excellence for Data Driven Discovery, Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory of Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory of Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Anna Worthmann
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Magnus B F Gustafsson
- Embark Laboratories, Copenhagen, Denmark
- Chemical Process Research and Development, Chemical Process Research & DevelopmentLEO Pharma, Ballerup, Denmark
| | - Mads Tang-Christensen
- Embark Laboratories, Copenhagen, Denmark
- School of Biomedical Sciences Faculty of Medicine, Nursing and Health Sciences Monash University, Melbourne, Victoria, Australia
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Paul Kievit
- Division of Metabolic Health and Disease, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Thue W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Embark Laboratories, Copenhagen, Denmark
| | - Jakob B Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
- Embark Laboratories, Copenhagen, Denmark.
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
- Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark.
- Embark Laboratories, Copenhagen, Denmark.
| |
Collapse
|
13
|
Nissan N, Allen MC, Sabatino D, Biggar KK. Future Perspective: Harnessing the Power of Artificial Intelligence in the Generation of New Peptide Drugs. Biomolecules 2024; 14:1303. [PMID: 39456236 PMCID: PMC11505729 DOI: 10.3390/biom14101303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
The expansive field of drug discovery is continually seeking innovative approaches to identify and develop novel peptide-based therapeutics. With the advent of artificial intelligence (AI), there has been a transformative shift in the generation of new peptide drugs. AI offers a range of computational tools and algorithms that enables researchers to accelerate the therapeutic peptide pipeline. This review explores the current landscape of AI applications in peptide drug discovery, highlighting its potential, challenges, and ethical considerations. Additionally, it presents case studies and future prospectives that demonstrate the impact of AI on the generation of new peptide drugs.
Collapse
Affiliation(s)
- Nour Nissan
- Institute of Biochemistry, Departments of Biology & Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada (D.S.)
- NuvoBio Corporation, Ottawa, ON K1S 5B6, Canada
| | - Mitchell C. Allen
- Institute of Biochemistry, Departments of Biology & Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada (D.S.)
| | - David Sabatino
- Institute of Biochemistry, Departments of Biology & Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada (D.S.)
| | - Kyle K. Biggar
- Institute of Biochemistry, Departments of Biology & Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada (D.S.)
- NuvoBio Corporation, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
14
|
Tsuji T, Inazuki H, Kobayashi D, Hayashi J, Denda M, Otaka A. Cysteinylprolyl ester-mediated drug release from a lipid-drug conjugate. Bioorg Med Chem Lett 2024; 109:129850. [PMID: 38879090 DOI: 10.1016/j.bmcl.2024.129850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/23/2024]
Abstract
For small-molecule drugs, lipidation via a cleavable linkage can extend half-life in circulation through interaction with albumin. Here we modified the cysteinylprolyl ester (CPE) system used in peptide thioester synthesis, which normally requires basic conditions, for use as an self-immolative linker and release device for a lipid-gemcitabine conjugate. To improve release under physiological conditions for medical application, a methyl group at the α-position of cysteine on the CPE unit was incorporated in anticipation of the Thorpe-Ingold effect. As a result, Ac-Gly-(α-Me)Cys(SH)-Pro-gemcitabine 11 drastically promoted the release of gemcitabine in comparison with Ac-Gly-Cys(SH)-Pro-gemcitabine 10. Furthermore, in the presence of bovine serum albumin and/or 2-mercaptoethanesulfonic acid, the gentle and continuous release of gemcitabine from the lipid-gemcitabine conjugate 16 was achieved. In addition to gemcitabine, this method could allow high clearance drugs, including nucleic acid and prostacyclin derivatives, to maintain their biological activity long enough to become effective.
Collapse
Affiliation(s)
- Takashi Tsuji
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Hayato Inazuki
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Daishiro Kobayashi
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Junya Hayashi
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Masaya Denda
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Akira Otaka
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan.
| |
Collapse
|
15
|
Zaykov AN, Gelfanov VM, Tagmose TM, Demozay D, Manfè V, Rohlfs R, Rivir M, Perez-Tilve D, Finan B, DiMarchi RD. Toward once-monthly insulin therapy via synergy in two pharmacokinetic protractors: Fc-conjugation and fatty acid acylation. RSC Chem Biol 2024; 5:763-775. [PMID: 39092439 PMCID: PMC11289878 DOI: 10.1039/d4cb00078a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/17/2024] [Indexed: 08/04/2024] Open
Abstract
Pharmacokinetic properties and duration of therapeutic action of a pharmaceutical agent can be significantly extended through the combination of two distinct strategies aimed at increasing plasma half-life: fatty acid acylation and Fc-conjugation. Using insulin as a case study, we demonstrate that a doubly protracted insulin analog produces a substantial prolongation of pharmacodynamic effect to lower blood glucose in STZ-treated mice when compared to the Fc-only counterparts. This enhancement is further corroborated by direct pharmacokinetic measurements in rat and dog models, demonstrating the potential for once-monthly insulin therapy. The results suggest that this approach might have broad application across a diverse spectrum of peptide- and protein-based therapeutics.
Collapse
Affiliation(s)
| | | | - Tina M Tagmose
- Novo Nordisk, Global Research Technologies DK-2760 Maaloev Denmark
| | - Damien Demozay
- Novo Nordisk, Global Research Technologies DK-2760 Maaloev Denmark
| | - Valentina Manfè
- Novo Nordisk, Global Research Technologies DK-2760 Maaloev Denmark
| | - Rebecca Rohlfs
- Novo Nordisk Research Center Indianapolis Indianapolis IN 46241 USA
| | - Marita Rivir
- Department of Pharmacology and Systems Physiology, University of Cincinnati-College of Medicine Cincinnati OH 45267 USA
| | - Diego Perez-Tilve
- Department of Pharmacology and Systems Physiology, University of Cincinnati-College of Medicine Cincinnati OH 45267 USA
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis Indianapolis IN 46241 USA
| | - Richard D DiMarchi
- Novo Nordisk Research Center Indianapolis Indianapolis IN 46241 USA
- Department of Chemistry, Indiana University Bloomington IN 47405 USA
| |
Collapse
|
16
|
Reddiar SB, Abdallah M, Styles IK, Müllertz OO, Trevaskis NL. Lymphatic uptake of the lipidated and non-lipidated GLP-1 agonists liraglutide and exenatide is similar in rats. Eur J Pharm Biopharm 2024; 200:114339. [PMID: 38789061 DOI: 10.1016/j.ejpb.2024.114339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/02/2024] [Accepted: 05/22/2024] [Indexed: 05/26/2024]
Abstract
Peptides, despite their therapeutic potential, face challenges with undesirable pharmacokinetic (PK) properties and biodistribution, including poor oral absorption and cellular uptake, and short plasma elimination half-lives. Lipidation of peptides is a common strategy to improve their physicochemical and PK properties, making them viable drug candidates. For example, the plasma half-life of peptides has been extended via conjugation to lipids that are proposed to promote binding to serum albumin and thus protect against rapid clearance. Recent work has shown that lipid conjugation to oligodeoxynucleotides, polymers and small molecule drugs results in association not only with albumin, but also with lipoproteins, resulting in half-life prolongation and transport from administration sites via the lymphatics. Enhancing delivery into the lymph increases the efficacy of vaccines and therapeutics with lymphatic targets such as immunotherapies. In this study, the plasma PK, lymphatic uptake, and bioavailability of the glucagon-like peptide-1 (GLP-1) receptor agonist peptides, liraglutide (lipidated) and exenatide (non-lipidated), were investigated following subcutaneous (SC) administration to rats. As expected, liraglutide displayed an apparent prolonged plasma half-life (9.1 versus 1 h), delayed peak plasma concentrations and lower bioavailability (∼10 % versus ∼100 %) compared to exenatide after SC administration. The lymphatic uptake of both peptides was relatively low (<0.5 % of the dose) although lymph to plasma concentration ratios were greater than one for several early timepoints suggesting some direct uptake into lymph. The low lymphatic uptake may be due to the nature of the conjugated lipid (a single-chain C16 palmitic acid in liraglutide) but suggests that other peptides with similar lipid conjugations may also have relatively modest lymphatic uptake. If delivery to the lymph is desired, conjugation to more lipophilic moieties with higher albumin and/or lipoprotein binding efficiencies, such as diacylglycerols, may be appropriate.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia
| | - Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia
| | - Olivia O Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Australia.
| |
Collapse
|
17
|
Hoogenboezem EN, Patel SS, Lo JH, Cavnar AB, Babb LM, Francini N, Gbur EF, Patil P, Colazo JM, Michell DL, Sanchez VM, McCune JT, Ma J, DeJulius CR, Lee LH, Rosch JC, Allen RM, Stokes LD, Hill JL, Vickers KC, Cook RS, Duvall CL. Structural optimization of siRNA conjugates for albumin binding achieves effective MCL1-directed cancer therapy. Nat Commun 2024; 15:1581. [PMID: 38383524 PMCID: PMC10881965 DOI: 10.1038/s41467-024-45609-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 01/29/2024] [Indexed: 02/23/2024] Open
Abstract
The high potential of siRNAs to silence oncogenic drivers remains largely untapped due to the challenges of tumor cell delivery. Here, divalent lipid-conjugated siRNAs are optimized for in situ binding to albumin to improve pharmacokinetics and tumor delivery. Systematic variation of the siRNA conjugate structure reveals that the location of the linker branching site dictates tendency toward albumin association versus self-assembly, while the lipid hydrophobicity and reversibility of albumin binding also contribute to siRNA intracellular delivery. The lead structure increases tumor siRNA accumulation 12-fold in orthotopic triple negative breast cancer (TNBC) tumors over the parent siRNA. This structure achieves approximately 80% silencing of the anti-apoptotic oncogene MCL1 and yields better survival outcomes in three TNBC models than an MCL-1 small molecule inhibitor. These studies provide new structure-function insights on siRNA-lipid conjugate structures that are intravenously injected, associate in situ with serum albumin, and improve pharmacokinetics and tumor treatment efficacy.
Collapse
Affiliation(s)
- Ella N Hoogenboezem
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Shrusti S Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Justin H Lo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ashley B Cavnar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lauren M Babb
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Nora Francini
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Eva F Gbur
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Prarthana Patil
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Danielle L Michell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Violeta M Sanchez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua T McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jinqi Ma
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Carlisle R DeJulius
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Linus H Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jonah C Rosch
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ryan M Allen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Larry D Stokes
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jordan L Hill
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kasey C Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Rebecca S Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
18
|
Tian H, Chang M, Lyu Y, Dong N, Yu N, Yin T, Zhang Y, He H, Gou J, Tang X. Intramuscular injection of palmitic acid-conjugated Exendin-4 loaded multivesicular liposomes for long-acting and improving in-situ stability. Expert Opin Drug Deliv 2024; 21:169-185. [PMID: 38224039 DOI: 10.1080/17425247.2024.2305110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
BACKGROUND Exendin-4 (Ex4) is a promising drug for diabetes mellitus with a half-life of 2.4 h in human bodies. Besides, the Ex4 formulations currently employed in the clinic or under development have problems pertaining to stability. In this study, palmitic acid-modified Ex4 (Pal-Ex4) was prepared and purified to extend the half-life of Ex4. In addition, Pal-Ex4-MVLs were further designed and optimized as a long-acting delivery system for intramuscular injection. METHODS Pal-Ex4 was encapsulated within multivesicular liposomes (MVLs) via a two-step double emulsification process. The formulated products were then assessed for their vesicle size, encapsulation efficiency, and in vitro and in vivo. RESULTS Pal-Ex4-MVLs with a notable encapsulation efficiency of 99.18% were successfully prepared. Pal-Ex4-MVLs, administered via a single intramuscular injection in Sprague-Dawley rats, sustained stable plasma concentrations for 168 h, presenting extended half-life (77.28 ± 12.919 h) and enhanced relative bioavailability (664.18%). MVLs protected Ex4 through providing stable retention and slow release. This approach considerably improved the in-situ stability of the drug for intramuscular administration. CONCLUSIONS The combination of palmitic acid modification process with MVLs provides dual protection for Ex4 and can be a promising strategy for other hydrophilic protein/polypeptide-loaded sustained-release delivery systems with high drug bioactivity.
Collapse
Affiliation(s)
- Huixian Tian
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Minsi Chang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Yanlin Lyu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Nan Dong
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Nini Yu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| |
Collapse
|
19
|
Abubakar M, Nama L, Ansari MA, Ansari MM, Bhardwaj S, Daksh R, Syamala KLV, Jamadade MS, Chhabra V, Kumar D, Kumar N. GLP-1/GIP Agonist as an Intriguing and Ultimate Remedy for Combating Alzheimer's Disease through its Supporting DPP4 Inhibitors: A Review. Curr Top Med Chem 2024; 24:1635-1664. [PMID: 38803170 DOI: 10.2174/0115680266293416240515075450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a widespread neurological illness in the elderly, which impacted about 50 million people globally in 2020. Type 2 diabetes has been identified as a risk factor. Insulin and incretins are substances that have various impacts on neurodegenerative processes. Preclinical research has shown that GLP-1 receptor agonists decrease neuroinflammation, tau phosphorylation, amyloid deposition, synaptic function, and memory formation. Phase 2 and 3 studies are now occurring in Alzheimer's disease populations. In this article, we present a detailed assessment of the therapeutic potential of GLP-1 analogues and DPP4 inhibitors in Alzheimer's disease. AIM This study aimed to gain insight into how GLP-1 analogues and associated antagonists of DPP4 safeguard against AD. METHODS This study uses terms from search engines, such as Scopus, PubMed, and Google Scholar, to explore the role, function, and treatment options of the GLP-1 analogue for AD. RESULTS The review suggested that GLP-1 analogues may be useful for treating AD because they have been linked to anti-inflammatory, neurotrophic, and neuroprotective characteristics. Throughout this review, we discuss the underlying causes of AD and how GLP signaling functions. CONCLUSION With a focus on AD, the molecular and pharmacological effects of a few GLP-1/GIP analogs, both synthetic and natural, as well as DPP4 inhibitors, have been mentioned, which are in the preclinical and clinical studies. This has been demonstrated to improve cognitive function in Alzheimer's patients.
Collapse
Affiliation(s)
- Mohammad Abubakar
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Lokesh Nama
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Mohammad Arif Ansari
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Mohammad Mazharuddin Ansari
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Shivani Bhardwaj
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Rajni Daksh
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Katta Leela Venkata Syamala
- Department of Regulatory and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Mohini Santosh Jamadade
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Vishal Chhabra
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Dileep Kumar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
- Department of Entomology, University of California, Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| |
Collapse
|
20
|
Gomez-Soler M, Olson EJ, de la Torre ER, Zhao C, Lamberto I, Flood DT, Danho W, Lechtenberg BC, Riedl SJ, Dawson PE, Pasquale EB. Lipidation and PEGylation Strategies to Prolong the in Vivo Half-Life of a Nanomolar EphA4 Receptor Antagonist. Eur J Med Chem 2023; 262:115876. [PMID: 38523699 PMCID: PMC10959496 DOI: 10.1016/j.ejmech.2023.115876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/08/2023] [Accepted: 10/12/2023] [Indexed: 03/26/2024]
Abstract
The EphA4 receptor tyrosine kinase plays a role in neurodegenerative diseases, inhibition of nerve regeneration, cancer progression and other diseases. Therefore, EphA4 inhibition has potential therapeutic value. Selective EphA4 kinase inhibitors are not available, but we identified peptide antagonists that inhibit ephrin ligand binding to EphA4 with high specificity. One of these peptides is the cyclic APY-d3 (βAPYCVYRβASWSC-NH2), which inhibits ephrin-A5 ligand binding to EphA4 with low nanomolar binding affinity and is highly protease resistant. Here we describe modifications of APY-d3 that yield two different key derivatives with greatly increased half-lives in the mouse circulation, the lipidated APY-d3-laur8 and the PEGylated APY-d3-PEG4. These two derivatives inhibit ligand induced EphA4 activation in cells with sub-micromolar potency. Since they retain high potency and specificity for EphA4, lipidated and PEGylated APY-d3 derivatives represent new tools for discriminating EphA4 activities in vivo and for preclinical testing of EphA4 inhibition in animal disease models.
Collapse
Affiliation(s)
- Maricel Gomez-Soler
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Erika J. Olson
- Departments of Chemistry and Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Elena Rubio de la Torre
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Chunxia Zhao
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Ilaria Lamberto
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Dillon T. Flood
- Departments of Chemistry and Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Waleed Danho
- Del Mar, California 92014, United States
- Deceased
| | - Bernhard C. Lechtenberg
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Stefan J. Riedl
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Philip E. Dawson
- Departments of Chemistry and Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Elena B. Pasquale
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| |
Collapse
|
21
|
Pilz M, Cavelius P, Qoura F, Awad D, Brück T. Lipopeptides development in cosmetics and pharmaceutical applications: A comprehensive review. Biotechnol Adv 2023; 67:108210. [PMID: 37460047 DOI: 10.1016/j.biotechadv.2023.108210] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/25/2023]
Abstract
Lipopeptides are surface active, natural products of bacteria, fungi and green-blue algae origin, having diverse structures and functionalities. In analogy, a number of chemical synthesis techniques generated new designer lipopeptides with desirable features and functions. Lipopetides are self-assembly guided, supramolecular compounds which have the capacity of high-density presentation of the functional epitopes at the surface of the nanostructures. This feature contributes to their successful application in several industry sectors, including food, feed, personal care, and pharmaceutics. In this comprehensive review, the novel class of ribosomally synthesized lipopeptides is introduced alongside the more commonly occuring non-ribosomal lipopeptides. We highlight key representatives of the most researched as well as recently described lipopeptide families, with emphasis on structural features, self-assembly and associated functions. The common biological, chemical and hybrid production routes of lipopeptides, including prominent analogues and derivatives are also discussed. Furthermore, genetic engineering strategies aimed at increasing lipopeptide yields, diversity and biological activity are summarized and exemplified. With respect to application, this work mainly details the potential of lipopeptides in personal care and cosmetics industry as cleansing agents, moisturizer, anti-aging/anti-wrinkling, skin whitening and preservative agents as well as the pharmaceutical industry as anitimicrobial agents, vaccines, immunotherapy, and cancer drugs. Given that this review addresses human applications, we conclude on the topic of safety of lipopeptide formulations and their sustainable production.
Collapse
Affiliation(s)
- Melania Pilz
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany
| | - Philipp Cavelius
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany
| | - Farah Qoura
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany
| | - Dania Awad
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany.
| | - Thomas Brück
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany.
| |
Collapse
|
22
|
Hu H, Ransdell AS, Qu H, Durbin JD, Valenzuela FA, Hernandez-Buquer S, Gonciarz MD. Probing the Binding Mechanism of Acylated Peptides to Human Serum Albumin. ACS Chem Biol 2023; 18:1158-1167. [PMID: 37145869 DOI: 10.1021/acschembio.3c00018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Peptides represent an increasingly important class of pharmaceutical products. During the last decade or so, acylation with fatty acids has demonstrated considerable success in prolonging the circulating half-life of therapeutic peptides by exploiting the ability of fatty acids to reversibly bind to human serum albumin (HSA), thus significantly impacting their pharmacological profiles. Employing methyl-13C-labeled oleic acid or palmitic acid as probe molecules and exploiting HSA mutants designed to probe fatty acid binding, the signals in two-dimensional (2D) nuclear magnetic resonance (NMR) spectra corresponding to high-affinity fatty acid binding sites in HSA were assigned. Subsequently, using a set of selected acylated peptides, competitive displacement experiments by 2D NMR identified a primary fatty acid binding site in HSA utilized in acylated peptide binding. These results represent an important first step toward understanding the structural basis for acylated peptides binding to HSA.
Collapse
Affiliation(s)
- Haitao Hu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Anthony S Ransdell
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Hongchang Qu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Jim D Durbin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Francisco A Valenzuela
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Selene Hernandez-Buquer
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Malgorzata D Gonciarz
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| |
Collapse
|
23
|
Esposito S, Krick A, Pasquier O, Bonche F, Ingenito R, Magotti P, Bianchi E, Monteagudo E, Gallo M, Cicero DO, Orsatti L, Veneziano M, Caretti F, Mele R, Roversi D, Gennari N, Brasseur D, Gauzy-Lazo L, Duclos O, Mauriac C, Illiano S, Mallart S. Fatty acid acylated peptide therapeutics: discovery of omega-n oxidation of the lipid chain as a novel metabolic pathway in preclinical species. J Pharm Biomed Anal 2023; 227:115256. [PMID: 36764268 DOI: 10.1016/j.jpba.2023.115256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/11/2023] [Accepted: 01/15/2023] [Indexed: 01/19/2023]
Abstract
We recently described C18 fatty acid acylated peptides as a new class of potent long-lasting single-chain RXFP1 agonists that displayed relaxin-like activities in vivo. Early pharmacokinetics and toxicological studies of these stearic acid acylated peptides revealed a relevant oxidative metabolism occurring in dog and minipig, and also seen at a lower extent in monkey and rat. Mass spectrometry combined to NMR spectroscopy studies revealed that the oxidation occurred, unexpectedly, on the stearic acid chain at ω-1, ω-2 and ω-3 positions. Structure-metabolism relationship studies on acylated analogues with different fatty acids lengths (C15-C20) showed that the extent of oxidation was higher with longer chains. The oxidized metabolites could be generated in vitro using liver microsomes and engineered bacterial CYPs. These systems were correlating poorly with in vivo metabolism observed across species; however, the results suggest that this biotransformation pathway might be catalyzed by some unknown CYP enzymes.
Collapse
Affiliation(s)
- Simone Esposito
- Biodistribution, Biotransformation and Analytical Science Unit, Experimental Pharmacology Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy.
| | - Alain Krick
- DMPK France, Sanofi R&D, 1 rue Pierre Brossolette, 91385 Chilly Mazarin, France.
| | - Olivier Pasquier
- DMPK France, Sanofi R&D, 1 rue Pierre Brossolette, 91385 Chilly Mazarin, France
| | - Fabrice Bonche
- DMPK France, Sanofi R&D, 1 rue Pierre Brossolette, 91385 Chilly Mazarin, France
| | - Raffaele Ingenito
- Peptide Chemistry Unit, Peptides & Small Molecules R&D Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Paola Magotti
- Peptide Chemistry Unit, Peptides & Small Molecules R&D Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Elisabetta Bianchi
- Peptide Chemistry Unit, Peptides & Small Molecules R&D Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Edith Monteagudo
- Biodistribution, Biotransformation and Analytical Science Unit, Experimental Pharmacology Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy; PK/PD & Bioanalytics Unit, Experimental Pharmacology Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Mariana Gallo
- Structural Biology Unit, Computational Chemistry & Structural Biology Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Daniel Oscar Cicero
- Department of Chemical Science and Technology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Laura Orsatti
- PK/PD & Bioanalytics Unit, Experimental Pharmacology Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Maria Veneziano
- PK/PD & Bioanalytics Unit, Experimental Pharmacology Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Fulvia Caretti
- PK/PD & Bioanalytics Unit, Experimental Pharmacology Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Riccardo Mele
- Biodistribution, Biotransformation and Analytical Science Unit, Experimental Pharmacology Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Daniela Roversi
- Peptide Chemistry Unit, Peptides & Small Molecules R&D Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Nadia Gennari
- High Throughput Biology & Screening, Translational Research Department, IRBM Spa, Via Pontina Km 30 600, 00 071 Pomezia, Rome, Italy
| | - Denis Brasseur
- Integrated Drug Discovery, Sanofi R&D, 1 rue Pierre Brossolette, 91385 Chilly Mazarin, France
| | - Laurence Gauzy-Lazo
- Integrated Drug Discovery, Sanofi R&D, 1 rue Pierre Brossolette, 91385 Chilly Mazarin, France
| | - Olivier Duclos
- Integrated Drug Discovery, Sanofi R&D, 1 rue Pierre Brossolette, 91385 Chilly Mazarin, France
| | - Christine Mauriac
- DMPK France, Sanofi R&D, 1 rue Pierre Brossolette, 91385 Chilly Mazarin, France
| | - Stephane Illiano
- Investigative Toxicology, Sanofi R&D, 1 rue Pierre Brossolette, 91385 Chilly Mazarin, France
| | - Sergio Mallart
- Integrated Drug Discovery, Sanofi R&D, 1 rue Pierre Brossolette, 91385 Chilly Mazarin, France.
| |
Collapse
|
24
|
Zhang X, Cai Y, Yao Z, Chi H, Li Y, Shi J, Zhou Z, Sun L. Discovery of novel OXM-based glucagon-like peptide 1 (GLP-1)/glucagon receptor dual agonists. Peptides 2023; 161:170948. [PMID: 36646385 DOI: 10.1016/j.peptides.2023.170948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Novel glucagon receptor (GCGR) and glucagon-like peptide 1 receptor (GLP-1R) dual agonists are reported to have improved efficacy over GLP-1R mono-agonists in treating type 2 diabetes (T2DM) and obesity. Here, we describe the discovery of a novel oxyntomodulin (OXM) based GLP-1R/GCGR dual agonist with potent and balanced potency toward GLP-1R and GCGR. The lead peptide OXM-7 was obtained via stepwise rational design and long-acting modification. In ICR and db/db mice, OXM-7 exhibited prominent acute and long-acting hypoglycemic effects. In diet-induced obesity (DIO) mice, twice-daily administration of OXM-7 produced significant weight loss, normalized lipid metabolism, and improved glucose control. In DIO-nonalcoholic steatohepatitis (NASH) mice, OXM-7 treatment significantly reversed hepatic steatosis, and reduced serum and hepatic lipid levels. These preclinical data suggest the therapeutic potential of OXM-7 as a novel anti-diabetic, anti-steatotic and/or anti-obesity agent.
Collapse
Affiliation(s)
- Xiaolong Zhang
- Food and Pharmaceutical Research Institute, Jiangsu Food & Pharmaceutical Science College, Huaian 223003, Jiangsu, PR China
| | - Yuchen Cai
- School of Engineering, China Pharmaceutical University, Nanjing 210009, Jiangsu, PR China
| | - Zhihong Yao
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing 314001, Zhejiang, PR China
| | - Heng Chi
- Food and Pharmaceutical Research Institute, Jiangsu Food & Pharmaceutical Science College, Huaian 223003, Jiangsu, PR China
| | - Yan Li
- Food and Pharmaceutical Research Institute, Jiangsu Food & Pharmaceutical Science College, Huaian 223003, Jiangsu, PR China
| | - Jingjing Shi
- Food and Pharmaceutical Research Institute, Jiangsu Food & Pharmaceutical Science College, Huaian 223003, Jiangsu, PR China
| | - Zhongbo Zhou
- School of Pharmacy, Youjiang Medical University for Nationalities, 98 Chengxiang Road, Baise 533000, Guangxi, PR China.
| | - Lidan Sun
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing 314001, Zhejiang, PR China.
| |
Collapse
|
25
|
Hoogenboezem EN, Patel SS, Cavnar AB, Lo JH, Babb LM, Francini N, Patil P, Colazo JM, Michell DL, Sanchez VM, McCune JT, Ma J, DeJulius CR, Lee LH, Rosch JC, Allen RM, Stokes LD, Hill JL, Vickers KC, Cook RS, Duvall CL. Structural Optimization of siRNA Conjugates for Albumin Binding Achieves Effective MCL1-Targeted Cancer Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528574. [PMID: 36824780 PMCID: PMC9948981 DOI: 10.1101/2023.02.14.528574] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
The high potential for therapeutic application of siRNAs to silence traditionally undruggable oncogenic drivers remains largely untapped due to the challenges of tumor cell delivery. Here, siRNAs were optimized for in situ binding to albumin through C18 lipid modifications to improve pharmacokinetics and tumor delivery. Systematic variation of siRNA conjugates revealed a lead structure with divalent C18 lipids each linked through three repeats of hexaethylene glycol connected by phosphorothioate bonds. Importantly, we discovered that locating the branch site of the divalent lipid structure proximally (adjacent to the RNA) rather than at a more distal site (after the linker segment) promotes association with albumin, while minimizing self-assembly and lipoprotein association. Comparison to higher albumin affinity (diacid) lipid variants and siRNA directly conjugated to albumin underscored the importance of conjugate hydrophobicity and reversibility of albumin binding for siRNA delivery and bioactivity in tumors. The lead conjugate increased tumor siRNA accumulation 12-fold in orthotopic mouse models of triple negative breast cancer over the parent siRNA. When applied for silencing of the anti-apoptotic oncogene MCL-1, this structure achieved approximately 80% MCL1 silencing in orthotopic breast tumors. Furthermore, application of the lead conjugate structure to target MCL1 yielded better survival outcomes in three independent, orthotopic, triple negative breast cancer models than an MCL1 small molecule inhibitor. These studies provide new structure-function insights on optimally leveraging siRNA-lipid conjugate structures that associate in situ with plasma albumin for molecular-targeted cancer therapy.
Collapse
Affiliation(s)
| | - Shrusti S. Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Ashley B. Cavnar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Justin H. Lo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Lauren M. Babb
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Nora Francini
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Prarthana Patil
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN
| | | | - Violeta M. Sanchez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Joshua T. McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Jinqi Ma
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | | | | | - Jonah C. Rosch
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
| | - Ryan M. Allen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Larry D. Stokes
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Jordan L. Hill
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Kasey C. Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Rebecca S. Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| |
Collapse
|
26
|
Wang JY, Wang QW, Yang XY, Yang W, Li DR, Jin JY, Zhang HC, Zhang XF. GLP-1 receptor agonists for the treatment of obesity: Role as a promising approach. Front Endocrinol (Lausanne) 2023; 14:1085799. [PMID: 36843578 PMCID: PMC9945324 DOI: 10.3389/fendo.2023.1085799] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/02/2023] [Indexed: 02/04/2023] Open
Abstract
Obesity is a complex disease characterized by excessive fat accumulation which is caused by genetic, environmental and other factors. In recent years, there has been an increase in the morbidity, disability rate,and mortality due to obesity, making it great threat to people's health and lives, and increasing public health care expenses. Evidence from previous studies show that weight loss can significantly reduce the risk of obesity-related complications and chronic diseases. Diet control, moderate exercise, behavior modification programs, bariatric surgery and prescription drug treatment are the major interventions used to help people lose weight. Among them, anti-obesity drugs have high compliance rates and cause noticeable short-term effects in reducing obese levels. However, given the safety or effectiveness concerns of anti-obesity drugs, many of the currently used drugs have limited clinical use. Glucagon-like peptide-1 receptor (GLP-1R) agonists are a group of drugs that targets incretin hormone action, and its receptors are widely distributed in nerves, islets, heart, lung, skin, and other organs. Several animal experiments and clinical trials have demonstrated that GLP-1R agonists are more effective in treating or preventing obesity. Therefore, GLP-1R agonists are promising agents for the treatment of obese individuals. This review describes evidence from previous research on the effects of GLP-1R agonists on obesity. We anticipate that this review will generate data that will help biomedical researchers or clinical workers develop obesity treatments based on GLP-1R agonists.
Collapse
Affiliation(s)
- Jing-Yue Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Quan-Wei Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xin-Yu Yang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Dong-Rui Li
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jing-Yu Jin
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Hui-Cong Zhang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xian-Feng Zhang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
27
|
Sessa L, Concilio S, Fominaya J, Eletto D, Piotto S, Busquets X. A new serotonin 2A receptor antagonist with potential benefits in Non-Alcoholic Fatty Liver Disease. Life Sci 2023; 314:121315. [PMID: 36581095 DOI: 10.1016/j.lfs.2022.121315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/28/2022]
Abstract
Peripheral 5-hydroxytryptamine 2A receptor (5-HT2AR) could be a new pharmacological target for NASH, an evolution of NAFLD characterized by hepatic steatosis, cytoskeletal alterations, and hepatic inflammation that can arise with or without fibrosis. SJT4a is a synthetic β-carboline antagonist for 5-HT2AR developed by SJT molecular research to treat NASH. We performed a combined in silico/in vivo study on this potential drug to elucidate its activity and possible mechanism of action. The in silico protocol compares SJT4a with four known 5-HT2AR ligands with different activities (LSD, methiothepin, zotepine, risperidone). We performed molecular docking calculations, evaluation of binding energy by AI-based methods and Molecular Dynamics simulations of the five ligand-target complexes. Moreover, we used a pseudo-semantic analysis to evaluate the potential mechanism of action of SJT4a. In silico predictions and pseudo-semantic analysis suggested antagonistic activity for SJT4a. The in silico prediction was confirmed by [3H]-5HT radioligand binding together with SJT4a competition analysis in CHO-K1 cell cultures expressing 5-HT2AR. SJT4a was then tested in vivo. We investigated the effect of 8 weeks of treatment with SJT4A on metabolic parameters, liver pathology, NAFLD activity score, and fibrosis stage in male DIO-NASH C57BL/6 J mice diet-induced obesity fed with an obesogenic diet compared with DIO-NASH and LEAN-CHOW vehicles. In our tests, SJT4a showed intense activity in diminishing the most relevant hallmarks of NASH in the DIO-NASH mice model. We proposed a possible mode of action for SJT4a based on its 5-HT2AR antagonist activity.
Collapse
Affiliation(s)
- Lucia Sessa
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, Fisciano 84084, SA, Italy; Bionam Center for Biomaterials, University of Salerno, Via Giovanni Paolo II, 132, Fisciano 84084, SA, Italy
| | - Simona Concilio
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, Fisciano 84084, SA, Italy; Bionam Center for Biomaterials, University of Salerno, Via Giovanni Paolo II, 132, Fisciano 84084, SA, Italy
| | - Jesús Fominaya
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, 07122 Palma, Spain
| | - Daniela Eletto
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, Fisciano 84084, SA, Italy
| | - Stefano Piotto
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, Fisciano 84084, SA, Italy; Bionam Center for Biomaterials, University of Salerno, Via Giovanni Paolo II, 132, Fisciano 84084, SA, Italy.
| | - Xavier Busquets
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, 07122 Palma, Spain.
| |
Collapse
|
28
|
Kurtzhals P, Østergaard S, Nishimura E, Kjeldsen T. Derivatization with fatty acids in peptide and protein drug discovery. Nat Rev Drug Discov 2023; 22:59-80. [PMID: 36002588 DOI: 10.1038/s41573-022-00529-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 01/28/2023]
Abstract
Peptides and proteins are widely used to treat a range of medical conditions; however, they often have to be injected and their effects are short-lived. These shortcomings of the native structure can be addressed by molecular engineering, but this is a complex undertaking. A molecular engineering technology initially applied to insulin - and which has now been successfully applied to several biopharmaceuticals - entails the derivatization of peptides and proteins with fatty acids. Various protraction mechanisms are enabled by the specific characteristics and positions of the attached fatty acid. Furthermore, the technology can ensure a long half-life following oral administration of peptide drugs, can alter the distribution of peptides and may hold potential for tissue targeting. Due to the inherent safety and well-defined chemical nature of the fatty acids, this technology provides a versatile approach to peptide and protein drug discovery.
Collapse
|
29
|
Su Y, Zhang S, Wu Z, Liu W, Chen J, Deng F, Chen F, Zhu D, Hou K. Pharmacoeconomic analysis (CER) of Dulaglutide and Liraglutide in the treatment of patients with type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1054946. [PMID: 36755915 PMCID: PMC9899911 DOI: 10.3389/fendo.2023.1054946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
AIM To evaluate the treatment effect Fand pharmacoeconomic value of Dugaglutide in women with type 2 diabetes. METHODS Women (n=96) with type 2 diabetes recruited from June 2019 to December 2021 were randomized into two equal groups. The control group was treated with Liraglutide, and the observation group was treated with Dulaglutide, both for 24 weeks. The blood glucose levels, biochemical index, insulin resistance index (HOMA-IR), cost-effect ratio (CER), and drug safety were determined and compared between the two groups. RESULTS Blood glucose levels, the biochemical index, and HOMA-IR were lower in both groups after the treatment (P < 0.05), and there was no statistical difference in the blood glucose levels, biochemical index and HOMA-IR between the two groups (P > 0.05). The CER levels did not differ statistically between the two groups (P > 0.05). Both the cost and the incidence of drug side effects during solution injection were lower in the observation group than in the control group after 24 weeks of treatment (P < 0.05). CONCLUSION Both Dulaglutide and Liraglutide can reduce blood glucose levels, improve biochemical index, and HOMA-IR levels in women with type 2 diabetes. Dulaglutide is more cost-effective and safe. CLINICAL TRIAL REGISTRATION https://www.chictr.org.cn/index.aspx, identifier ChiCTR1900026514.
Collapse
Affiliation(s)
- Yu Su
- Center of Teaching Evaluation and Faculty Development, Anhui University of Chinese medicine, Hefei, Anhui, China
| | - Shuo Zhang
- Medical College of Shantou University, Shantou, China
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Zezhen Wu
- Medical College of Shantou University, Shantou, China
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Weiting Liu
- School of nursing, Anhui University of Chinese medicine, Hefei, Anhui, China
| | - Jingxian Chen
- Medical College of Shantou University, Shantou, China
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Feiying Deng
- Medical College of Shantou University, Shantou, China
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Fengwu Chen
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Dan Zhu
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Kaijian Hou
- School of Public Health, Shantou University, Shantou, China
- *Correspondence: Kaijian Hou,
| |
Collapse
|
30
|
Douton JE, Acharya NK, Stoltzfus B, Sun D, Grigson PS, Nyland JE. Acute glucagon-like peptide-1 receptor agonist liraglutide prevents cue-, stress-, and drug-induced heroin-seeking in rats. Behav Pharmacol 2022; 33:364-378. [PMID: 35695511 PMCID: PMC9308649 DOI: 10.1097/fbp.0000000000000685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Substance use disorder is challenging to treat due to its relapsing nature. In the last decade, opioid use disorder has been a threat to public health, being declared an epidemic by the Centers for Disease Control and Prevention. This is a tragic situation, considering there currently are only three effective, yet not ideal, treatments to prevent relapse to opioids. Recent research has shown that hormones that modulate hunger and satiety also can modulate motivated behavior for drugs of abuse. For example, the short-acting analog of glucagon-like peptide-1 (GLP-1), an incretin hormone that regulates homeostatic feeding, has been shown to reduce responding for rewarding stimuli such as food, cocaine, heroin, and nicotine when administered over several days or weeks. This may serve as an effective adjuvant during treatment; however, whether it would be effective when used acutely to bridge a patient between cessation of use and onset of medication for the treatment of an opioid addiction is unknown. Here, we tested the acute effects of the longer acting GLP-1 analog, liraglutide, on heroin-seeking. In rats with heroin self-administration experience, we found that subcutaneous administration of an acute dose of 0.3-mg/kg liraglutide was effective in preventing drug-seeking after exposure to three major precipitators: drug-associated cues, stress (yohimbine-induced), and the drug itself. Finally, we confirmed that the reduction in drug-seeking is not due to a locomotor impairment, as liraglutide did not significantly alter performance in a rotarod test. As such, acute use of GLP-1 analogs may serve as a new and effective nonopioid bridge to treatment.
Collapse
Affiliation(s)
- Joaquin E Douton
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nikhil K Acharya
- Department of Neural and Behavioral Sciences, Penn State College of Medicine
| | - Brooke Stoltzfus
- Department of Neural and Behavioral Sciences, Penn State College of Medicine
| | - Dongxiao Sun
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Patricia S Grigson
- Department of Neural and Behavioral Sciences, Penn State College of Medicine
| | - Jennifer E Nyland
- Department of Neural and Behavioral Sciences, Penn State College of Medicine
| |
Collapse
|
31
|
Yang B, Gomes Dos Santos A, Puri S, Bak A, Zhou L. The industrial design, translation, and development strategies for long-acting peptide delivery. Expert Opin Drug Deliv 2022; 19:1233-1245. [PMID: 35787229 DOI: 10.1080/17425247.2022.2098276] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Peptides are widely recognized as therapeutic agents in the treatment of a wide range of diseases, such as cancer, diabetes etc. However, their use has been limited by their short half-life, due to significant metabolism by exo- and endo-peptidases as well as their inherent poor physical and chemical stability. Research with the aim of improving their half-life in the body, and thus improving patient compliance (by decreasing the frequency of injections) has gained significant attention. AREAS COVERED This review outlines the current landscape and industrial approaches to achieve extended peptide exposure and reduce dosing frequency. Emphasis is placed on identifying challenges in drug product manufacturing and desirable critical quality attributes that are essential for activity and safety, providing insights into chemistry and design aspects impacting peptide release, and summarizing important considerations for CMC developability assessments of sustained release peptide drugs. EXPERT OPINION Bring the patient and disease perspective early into development. Substantial advances have been made in the field of sustained delivery of peptides despite their complexity. The article will also highlight considerations for early-stage product design and development, providing an industrial perspective on risk mitigation in developing sustained release peptide drug products.
Collapse
Affiliation(s)
- Bin Yang
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Ana Gomes Dos Santos
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Sanyogitta Puri
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Annette Bak
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, USA
| | - Liping Zhou
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, USA
| |
Collapse
|
32
|
Klepach A, Tran H, Ahmad Mohammed F, ElSayed ME. Characterization and impact of peptide physicochemical properties on oral and subcutaneous delivery. Adv Drug Deliv Rev 2022; 186:114322. [PMID: 35526665 DOI: 10.1016/j.addr.2022.114322] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/21/2022] [Accepted: 05/02/2022] [Indexed: 11/25/2022]
Abstract
Peptides, an emerging modality within the biopharmaceutical industry, are often delivered subcutaneously with evolving prospects on oral delivery. Barrier biology within the subcutis or gastrointestinal tract is a significant challenge in limiting absorption or otherwise disrupting peptide disposition. Aspects of peptide pharmacokinetic performance and ADME can be mitigated with careful molecular design that tailors for properties such as effective size, hydrophobicity, net charge, proteolytic stability, and albumin binding. In this review, we endeavor to highlight effective techniques in qualifying physicochemical properties of peptides and discuss advancements of in vitro models of subcutaneous and oral delivery. Additionally, we will delineate empirical findings around the relationship of these physicochemical properties and in vivo (animal or human) impact. We conclude that robust peptide characterization methods and in vitro techniques with demonstrated correlations to in vivo data are key routines to incorporate in the drug discovery and development to improve the probability of technical and commercial success of peptide therapeutics.
Collapse
|
33
|
Chen H, Lu Y, Shi S, Zhang Q, Cao X, Sun L, An D, Zhang X, Kong X, Liu J. Design and Development of a New Glucagon-Like Peptide-1 Receptor Agonist to Obtain High Oral Bioavailability. Pharm Res 2022; 39:1891-1906. [PMID: 35698011 DOI: 10.1007/s11095-022-03265-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/18/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Semaglutide is the only oral GLP-1 RA in the market, but oral bioavailability is generally limited in range of 0.4-1%. In this study, a new GLP-1RA named SHR-2042 was developed to gain higher oral bioavailability than semaglutide. METHOD Self-association of SHR-2042, semaglutide and liraglutide were assessed using SEC-MALS. The intestinal perfusion test in SD rats was used to select permeation enhancers (PEs) including SNAC, C10 and LCC. ITC, CD and DLS were used to explore the interaction between SHR-2042 and SNAC. Gastric administrated test in SD rats was used to screen SHR-2042 granules with different SHR-2042/SNAC ratios. The oral bioavailability of SHR-2042 was studied in rats and monkeys. RESULT The designed GLP-1RA, SHR-2042, gives a better solubility and lipophilicity than semaglutide. While it forms a similar oligomer with that of semaglutide. During the selection of PEs, SNAC shows better exposure than the other competing PEs including C10 and LCC. SHR-2042 and SNAC bind quickly and exhibit hydrophobic interaction. SNAC could promote monomerization of SHR-2042 and form micelles to trap the monomerized SHR-2042. The oral bioavailability of SHR-2042 paired with SNAC is 0.041% (1:0, w/w), 0.083% (1:10, w/w), 0.32% (1:30, w/w) and 2.83% (1:60, w/w) in rats. And the oral bioavailability of SHR-2042 matched with SNAC is 3.39% (1:30, w/w) in monkeys, which is over 10 times higher than that of semaglutide. CONCLUSION We believe that the design and development of oral SHR-2042 will provide a new way to design more and more GLP-1RAs with high oral bioavailability in the future.
Collapse
Affiliation(s)
- Hao Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Yun Lu
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Shuai Shi
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Qiang Zhang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Xiaoli Cao
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Lei Sun
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Dong An
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Xiaojie Zhang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Xianglin Kong
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Jianping Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
34
|
Sun L, Zheng ZM, Shao CS, Zhang ZY, Li MW, Wang L, Wang H, Zhao GH, Wang P. Rational Design by Structural Biology of Industrializable, Long-Acting Antihyperglycemic GLP-1 Receptor Agonists. Pharmaceuticals (Basel) 2022; 15:ph15060740. [PMID: 35745659 PMCID: PMC9230455 DOI: 10.3390/ph15060740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is easily degraded by dipeptidyl peptidase-4 (DPP-4) in the human body, limiting its therapeutic effect on type II diabetes. Therefore, improving GLP-1 receptor agonist (GLP-1RA) stability is a major obstacle for drug development. We analyzed human GLP-1, DPP-4, and GLP-1 receptor structures and designed three GLP-1RAs, which were introduced into fusion protein fragments and changed in the overall conformation. This modification effectively prevented GLP-1RAs from entering the DPP-4 active center without affecting GLP-1RAs’ ability to bind to GLP-1R, the new GLP-1RA hypoglycemic effect lasting for >24 h. Through molecular modeling, molecular dynamics calculation, and simulation, possible tertiary structure models of GLP-1RAs were obtained; molecular docking with DPP-4 and GLP-1R showed access to the fusion protein. The overall conformational change of GLP-1RAs prevented DPP-4 binding, without affecting GLP-1RAs’ affinity to GLP-1R. This study provides important drug design ideas for GLP-1RA development and a new example for application of structural biology-based protein design in drug development.
Collapse
Affiliation(s)
- Lei Sun
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China; (L.S.); (C.-S.S.); (L.W.); (H.W.); (G.-H.Z.)
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China
| | - Zhi-Ming Zheng
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China; (L.S.); (C.-S.S.); (L.W.); (H.W.); (G.-H.Z.)
- Correspondence: (Z.-M.Z.); (P.W.); Tel./Fax: +86-551-65593148 (Z.-M.Z.); +86-551-65593145 (P.W.)
| | - Chang-Sheng Shao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China; (L.S.); (C.-S.S.); (L.W.); (H.W.); (G.-H.Z.)
| | - Zhi-Yong Zhang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, National Science Center for Physical Sciences at Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230022, China; (Z.-Y.Z.); (M.-W.L.)
| | - Ming-Wei Li
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, National Science Center for Physical Sciences at Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230022, China; (Z.-Y.Z.); (M.-W.L.)
| | - Li Wang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China; (L.S.); (C.-S.S.); (L.W.); (H.W.); (G.-H.Z.)
| | - Han Wang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China; (L.S.); (C.-S.S.); (L.W.); (H.W.); (G.-H.Z.)
| | - Gen-Hai Zhao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China; (L.S.); (C.-S.S.); (L.W.); (H.W.); (G.-H.Z.)
| | - Peng Wang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China; (L.S.); (C.-S.S.); (L.W.); (H.W.); (G.-H.Z.)
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China
- Correspondence: (Z.-M.Z.); (P.W.); Tel./Fax: +86-551-65593148 (Z.-M.Z.); +86-551-65593145 (P.W.)
| |
Collapse
|
35
|
Frimann TM, Ko SK, Harris P, Bukrinski JT, Peters GHJ. In-silico study of the interactions between acylated glucagon like-peptide-1 analogues and the native receptor. J Biomol Struct Dyn 2022:1-15. [PMID: 35612899 DOI: 10.1080/07391102.2022.2078409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We have performed a series of multiple molecular dynamics (MD) simulations of glucagon-like peptide-1 (GLP-1) and acylated GLP-1 analogues in complex with the endogenous receptor (GLP-1R) to obtain a molecular understanding of how fatty acid (FA) chain structure, acylation position on the peptide, and presence of a linker affect the binding. MD simulations were analysed to extract heatmaps of receptor-peptide interaction patterns and to determine the free energy of binding using the molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) approach. The extracted free energies from MM-PBSA calculations are in qualitative agreement with experimentally determined potencies. Furthermore, the interaction patterns seen in the receptor-GLP-1 complex simulations resemble previously reported binding interactions validating the simulations. Analysing the receptor-GLP-1 analogue complex simulations, we found that the major differences between the systems stem from FA interactions and positioning of acylation in the peptide. Hydrophobic interactions between the FA chain and a hydrophobic patch on the extracellular domain contribute significantly to the binding affinity. Acylation on Lys26 resulted in noticeably more interactions between the FA chain and the extracellular domain hydrophobic patch than found for acylation on Lys34 and Lys38, respectively. The presence of a charged linker between the peptide and FA chain can potentially stabilise the complex by forming hydrogen bonds to arginine residues in the linker region between the extracellular domain and the transmembrane domain. A molecular understanding of the fatty acid structure and its effect on binding provides important insights into designing acylated agonists for GLP-1R.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Tine Maja Frimann
- Department of Chemistry, Technical University of Denmark, Lyngby, Denmark
| | - Suk Kyu Ko
- Department of Chemistry, Technical University of Denmark, Lyngby, Denmark
| | - Pernille Harris
- Department of Chemistry, Technical University of Denmark, Lyngby, Denmark.,Department of Chemistry, H.C. Ørsted Institute, University of Copenhagen, Copenhagen, Denmark
| | | | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
36
|
Sun X, Zhang Z, Liu M, Zhang P, Nie L, Liu Y, Chen Y, Xu F, Liu Z, Zeng Y. Small-molecule albumin ligand modification to enhance the anti-diabetic ability of GLP-1 derivatives. Biomed Pharmacother 2022; 148:112722. [DOI: 10.1016/j.biopha.2022.112722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 11/02/2022] Open
|
37
|
Lipidated PrRP31 metabolites are long acting dual GPR10 and NPFF2 receptor agonists with potent body weight lowering effect. Sci Rep 2022; 12:1696. [PMID: 35105898 PMCID: PMC8807614 DOI: 10.1038/s41598-022-05310-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 01/11/2022] [Indexed: 12/16/2022] Open
Abstract
Prolactin-releasing peptide (PrRP) is an endogenous neuropeptide involved in appetite regulation and energy homeostasis. PrRP binds with high affinity to G-protein coupled receptor 10 (GPR10) and with lesser activity towards the neuropeptide FF receptor type 2 (NPFF2R). The present study aimed to develop long-acting PrRP31 analogues with potent anti-obesity efficacy. A comprehensive series of C18 lipidated PrRP31 analogues was characterized in vitro and analogues with various GPR10 and NPFF2R activity profiles were profiled for bioavailability and metabolic effects following subcutaneous administration in diet-induced obese (DIO) mice. PrRP31 analogues acylated with a C18 lipid chain carrying a terminal acid (C18 diacid) were potent GPR10-selective agonists and weight-neutral in DIO mice. In contrast, acylation with aliphatic C18 lipid chain (C18) resulted in dual GPR10-NPFF2R co-agonists that suppressed food intake and promoted a robust weight loss in DIO mice, which was sustained for at least one week after last dosing. Rapid in vivo degradation of C18 PrRP31 analogues gave rise to circulating lipidated PrRP metabolites maintaining dual GPR10-NPFF2R agonist profile and long-acting anti-obesity efficacy in DIO mice. Combined GPR10 and NPFF2R activation may therefore be a critical mechanism for obtaining robust anti-obesity efficacy of PrRP31 analogues.
Collapse
|
38
|
Zhang P, Xu M, Ding J, Chen J, Zhang T, Huo L, Liu Z. Fatty acid-conjugated radiopharmaceuticals for fibroblast activation protein-targeted radiotherapy. Eur J Nucl Med Mol Imaging 2021; 49:1985-1996. [PMID: 34746969 DOI: 10.1007/s00259-021-05591-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/11/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Radiopharmaceuticals that target cancer-associated fibroblasts (CAFs) have become an increasingly attractive strategy for cancer theranostics. Recently, a series of fibroblast activation protein inhibitor (FAPI)-based radiopharmaceuticals have been successfully applied to the diagnosis of a variety of cancers and exhibited excellent tumor selectivity. Nevertheless, CAF-targeted radionuclide therapy encounters difficulties in cancer treatment, as the tumor uptake and retention of FAPIs are insufficient. To meet this challenge, we tried to conjugate albumin-binding moiety to FAPI molecule for prolonged circulation that may increase the accumulation and retention of radiopharmaceuticals in tumor. METHODS Two fatty acids, lauric acid (C12) and palmitic acid (C16), were conjugated to FAPI-04 to give two albumin-binding FAPI radiopharmaceuticals, denoted as FAPI-C12 and FAPI-C16, respectively. They had been radiolabeled with gallium-68, yttrium-86, and lutecium-177 for stability study, binding affinity assay, PET and SPECT imaging, biodistribution, and radionuclide therapy study to systematically evaluate their potential for CAF-targeted radionuclide therapy. RESULTS FAPI-C12 and FAPI-C16 showed high binding affinity to FAP with the IC50 of 6.80 ± 0.58 nM and 5.06 ± 0.69 nM, respectively. They were stable in both saline and plasma. The tumor uptake of [68Ga]Ga-FAPI-04 decreased by 56.9% until 30 h after treated with FAPI-C16 before, and the uptakes of [86Y]Y-FAPI-C12 and [86Y]Y-FAPI-C16 in HT-1080-FAP tumor were both much higher than that of HT-1080-Vehicle tumor which identified the high FAP specific of these two radiopharmaceuticals. Both FAPI-C12 and FAPI-C16 showed notably longer circulation and significantly enhanced tumor uptake than those of FAPI-04. [177Lu]Lu-FAPI-C16 had the higher tumor uptake at both 24 h (11.22 ± 1.18%IA/g) and 72 h (6.50 ± 1.19%IA/g) than that of [177Lu]Lu-FAPI-C12 (24 h, 7.54 ± 0.97%IA/g; 72 h, 2.62 ± 0.65%IA/g); both of them were much higher than [177Lu]Lu-FAPI-04 with the value of 1.24 ± 0.54%IA/g at 24 h after injection. Significant tumor volume inhibition of [177Lu]Lu-FAPI-C16 at the high activity of 29.6 MBq was observed, and the median survival was 28 days which was much longer than that of the [177Lu]Lu-FAPI-04 treated group of which the median survival was only 10 days. CONCLUSION This proof-of-concept study validates the hypothesis that conjugation of albumin binders may shift the pharmacokinetics and enhance the tumor uptake of FAPI-based radiopharmaceuticals. This could be a general strategy to transform the diagnostic FAP-targeted radiopharmaceuticals into their therapeutic pairs.
Collapse
Affiliation(s)
- Pu Zhang
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, 100871, Beijing, China
| | - Mengxin Xu
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, 100871, Beijing, China
| | - Jie Ding
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Junyi Chen
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, 100871, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| | - Zhibo Liu
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, 100871, Beijing, China.
- Peking University-Tsinghua University Center for Life Sciences, Beijing, 100871, China.
| |
Collapse
|
39
|
Østergaard S, Paulsson JF, Kofoed J, Zosel F, Olsen J, Jeppesen CB, Spetzler J, Ynddal L, Schleiss LG, Christoffersen BØ, Raun K, Sensfuss U, Nielsen FS, Jørgensen R, Wulff BS. The effect of fatty diacid acylation of human PYY 3-36 on Y 2 receptor potency and half-life in minipigs. Sci Rep 2021; 11:21179. [PMID: 34707178 PMCID: PMC8551270 DOI: 10.1038/s41598-021-00654-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/13/2021] [Indexed: 01/02/2023] Open
Abstract
Peptides are notoriously known to display very short in vivo half-lives often measured in minutes which in many cases greatly reduces or eliminates sufficient in vivo efficacy. To obtain long half-lives allowing for up to once-weekly dosing regimen, fatty acid acylation (lipidation) have been used to non-covalently associate the peptide to serum albumin thus serving as a circulating depot. This approach is generally considered in the scientific and patent community as a standard approach to protract almost any given peptide. However, it is not trivial to prolong the half-life of peptides by lipidation and still maintain high potency and good formulation properties. Here we show that attaching a fatty acid to the obesity-drug relevant peptide PYY3-36 is not sufficient for long pharmacokinetics (PK), since the position in the backbone, but also type of fatty acid and linker strongly influences PK and potency. Furthermore, understanding the proteolytic stability of the backbone is key to obtain long half-lives by lipidation, since backbone cleavage still occurs while associated to albumin. Having identified a PYY analogue with a sufficient half-life, we show that in combination with a GLP-1 analogue, liraglutide, additional weight loss can be achieved in the obese minipig model.
Collapse
Affiliation(s)
- Søren Østergaard
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark.
| | - Johan F Paulsson
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark
| | - Jacob Kofoed
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark
| | - Franziska Zosel
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark
| | - Jørgen Olsen
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark
| | - Claus Bekker Jeppesen
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark
| | - Jane Spetzler
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark
| | - Lars Ynddal
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark.,Gubra Aps, Hørsholm Kongevej 11B, 2970, Hørsholm, Denmark
| | - Luise Gram Schleiss
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark
| | | | - Kirsten Raun
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark
| | - Ulrich Sensfuss
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark.,STipe Therapeutics, Copenhagen, Denmark
| | - Flemming Seier Nielsen
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark
| | - Rasmus Jørgensen
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark.,CitoKi Pharma, Værløse, Denmark
| | - Birgitte S Wulff
- Global Research Technologies, Novo Nordisk A/S, Novo Nordisk Research Park, 2760, Maaloev, Denmark
| |
Collapse
|
40
|
Reiner J, Berlin P, Held J, Thiery J, Skarbaliene J, Griffin J, Russell W, Eriksson PO, Berner-Hansen M, Ehlers L, Vollmar B, Jaster R, Witte M, Lamprecht G. Dapiglutide, a novel dual GLP-1 and GLP-2 receptor agonist, attenuates intestinal insufficiency in a murine model of short bowel. JPEN J Parenter Enteral Nutr 2021; 46:1107-1118. [PMID: 34705281 DOI: 10.1002/jpen.2286] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Extensive intestinal resection may lead to short bowel (SB) syndrome, resulting in intestinal insufficiency or intestinal failure (IF). Intestinal insufficiency and IF involve deficiency of the proglucagon-derived hormones glucagon-like peptide-1 (GLP-1) and GLP-2. Two major problems of SB are epithelial surface loss and accelerated transit. Standard treatment now targets intestinal adaptation with a GLP-2 analogue to enlarge absorptive surface area. It is possible that additional benefit can be gained from a combination of GLP-1 and GLP-2 activity, with the aim to enlarge intestinal surface area and slow intestinal transit. METHODS The GLP-1- and GLP-2-specific effects of the novel dual GLP-1 receptor (GLP-1R) and GLP-2 receptor (GLP-2R) agonist dapiglutide (rINN) were characterized in rodents. Furthermore, in a murine SB model of intestinal insufficiency with 40% ileocecal resection, the influence of dapiglutide on intestinal growth, body weight, food intake, volume status, and stool water content was tested against vehicle and sham-operated male mice. RESULTS Dapiglutide significantly improves oral glucose tolerance, reduces intestinal transit time, and promotes intestinal growth. In the SB mouse model, dapiglutide promotes body weight recovery, despite unchanged intake of liquid diet. Dapiglutide promotes significant intestinal growth, as indicated by significantly increased villus height as well as intestinal length. Furthermore, dapiglutide reduces stool water losses, resulting in reduced plasma aldosterone. CONCLUSION Dapiglutide possesses specific and potent GLP-1R and GLP-2R agonist effects in rodents. In the murine SB model, combined unimolecular GLP-1R and GLP-2R stimulation with dapiglutide potently attenuates intestinal insufficiency and potentially also IF.
Collapse
Affiliation(s)
- Johannes Reiner
- Division of Gastroenterology and Endocrinology, Rostock University Medical Center, Rostock, 18057, Germany
| | - Peggy Berlin
- Division of Gastroenterology and Endocrinology, Rostock University Medical Center, Rostock, 18057, Germany
| | - Jascha Held
- Division of Gastroenterology and Endocrinology, Rostock University Medical Center, Rostock, 18057, Germany
| | - Johanna Thiery
- Division of Gastroenterology and Endocrinology, Rostock University Medical Center, Rostock, 18057, Germany
| | | | | | - Wayne Russell
- Research and Development, Zealand Pharma, Søborg, 2860, Denmark
| | | | | | - Luise Ehlers
- Division of Gastroenterology and Endocrinology, Rostock University Medical Center, Rostock, 18057, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Center, Rostock, 18057, Germany
| | - Robert Jaster
- Division of Gastroenterology and Endocrinology, Rostock University Medical Center, Rostock, 18057, Germany
| | - Maria Witte
- Department of General, Visceral, Thoracic, Vascular and Transplantat Surgery, Rostock University Medical Center, Rostock, 18057, Germany
| | - Georg Lamprecht
- Division of Gastroenterology and Endocrinology, Rostock University Medical Center, Rostock, 18057, Germany
| |
Collapse
|
41
|
Craig SL, Irwin N, Gault VA. Xenin and Related Peptides: Potential Therapeutic Role in Diabetes and Related Metabolic Disorders. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2021; 14:11795514211043868. [PMID: 34588834 PMCID: PMC8474313 DOI: 10.1177/11795514211043868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/09/2021] [Indexed: 11/16/2022]
Abstract
Xenin bioactivity and its role in normal physiology has been investigated by several research groups since its discovery in 1992. The 25 amino acid peptide hormone is secreted from the same enteroendocrine K-cells as the incretin hormone glucose-dependent insulinotropic polypeptide (GIP), with early studies highlighting the biological significance of xenin in the gastrointestinal tract, along with effects on satiety. Recently there has been more focus directed towards the role of xenin in insulin secretion and potential for diabetes therapies, especially through its ability to potentiate the insulinotropic actions of GIP as well as utilisation in dual/triple acting gut hormone therapeutic approaches. Currently, there is a lack of clinically approved therapies aimed at restoring GIP bioactivity in type 2 diabetes mellitus, thus xenin could hold real promise as a diabetes therapy. The biological actions of xenin, including its ability to augment insulin secretion, induce satiety effects, as well as restoring GIP sensitivity, earmark this peptide as an attractive antidiabetic candidate. This minireview will focus on the multiple biological actions of xenin, together with its proposed mechanism of action and potential benefits for the treatment of metabolic diseases such as diabetes.
Collapse
Affiliation(s)
- Sarah L Craig
- Faculty of Life and Health Sciences, School of Biomedical Sciences, Ulster University, UK
| | - Nigel Irwin
- Faculty of Life and Health Sciences, School of Biomedical Sciences, Ulster University, UK
| | - Victor A Gault
- Faculty of Life and Health Sciences, School of Biomedical Sciences, Ulster University, UK
| |
Collapse
|
42
|
Ruan S, Yang G, Dong Y, Shangguan W, Lu W. Discovery of a Long-Acting Parathyroid Hormone 1-34 Analogue to Treat Hypoparathyroidism. Mol Pharm 2021; 18:3260-3271. [PMID: 34482698 DOI: 10.1021/acs.molpharmaceut.1c00149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hypoparathyroidism (HP) is a rare disease with clinical manifestations of hypocalcemia and hyperphosphatemia, resulting from deficient or absent parathyroid hormone (PTH) secretion. Conventional treatment for patients with HP involves extensive calcium and vitamin D supplementation. In 2015, PTH1-84 was approved by the United States Food and Drug Administration as an adjunct for HP patients who cannot be well-controlled on conventional treatment. However, PTH1-84 therapy requires a daily injection, leading to poor patient compliance. The purpose of this study was to develop a long-acting PTH1-34 analogue by increasing its affinity to albumin. Three PTH1-34 variants were generated by substituting two of the three lysine (Lys) residues with arginine, reserving a single Lys as the modification site in each sequence. A series of side chains, containing fatty acid, deoxycholic acid, or biotin groups, were synthesized to modify these PTH1-34 variants by using a solid-liquid phase synthesis approach. In vitro bioactivity and albumin affinity tests were used to screen these new PTH1-34 analogues. Finally, Lys27-AAPC was selected from 69 synthesized analogues as a candidate therapeutic compound because it retained potency and exhibited a high albumin-binding capacity. In pharmacodynamic experiments, Lys27-AAPC demonstrated enhanced and prolonged efficacy in serum calcium elevating relative to PTH1-84. Moreover, a lyophilized powder for injection containing Lys27-AAPC was developed for further testing and represented a potential long-acting HP treatment.
Collapse
Affiliation(s)
- Sida Ruan
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Guiying Yang
- Shanghai Duomirui Biotechnology Ltd., Shanghai 201203, China
| | - Yuanzhen Dong
- Shanghai Duomirui Biotechnology Ltd., Shanghai 201203, China
| | - Wenwen Shangguan
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Weigen Lu
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| |
Collapse
|
43
|
Kalra S, Bhattacharya S, Kapoor N. Contemporary Classification of Glucagon-Like Peptide 1 Receptor Agonists (GLP1RAs). Diabetes Ther 2021; 12:2133-2147. [PMID: 34268675 PMCID: PMC8342688 DOI: 10.1007/s13300-021-01113-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/01/2021] [Indexed: 02/06/2023] Open
Abstract
This communication provides a contemporary classification of glucagon-like peptide 1 receptor agonists (GLP1RAs) based on indication, route, and frequency of administration, which could support a person-centric approach to treatment choice. It includes all recently developed GLP1RAs as well as those in advanced stages of clinical study. Keeping pace with current trends in pharmacology and metabolic medicine, it attempts to bring clarity and simplicity to a complex spread of information.
Collapse
Affiliation(s)
- Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, India
| | | | - Nitin Kapoor
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, India.
- Non Communicable Disease Unit, Melbourne School of Population and Global Health, University of Melbourne, Victoria, Australia.
| |
Collapse
|
44
|
Zhang L, Zhang L, Li Y, Li L, Melchiorsen JU, Rosenkilde M, Hölscher C. The Novel Dual GLP-1/GIP Receptor Agonist DA-CH5 Is Superior to Single GLP-1 Receptor Agonists in the MPTP Model of Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2021; 10:523-542. [PMID: 31958096 DOI: 10.3233/jpd-191768] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative disease for which there is no cure. In a clinical trial, the glucagon-like peptide-1 (GLP-1) receptor agonist exendin-4 has shown good protective effects in PD patients. The hormone glucose-dependent insulinotropic polypeptide (GIP) has also shown protective effects in animal models of PD. OBJECTIVE We tested DA-CH5, a novel dual GLP-1/GIP receptor agonist. METHODS DA-CH5 activity was tested on cells expressing GLP-1, GLP-2, GIP or glucagon receptors. The ability to cross the blood-brain barrier (BBB) of DA-CH5, exendin-4, liraglutide or other dual receptor agonists was tested with fluorescein-labelled peptides. DA-CH5, exendin-4 and liraglutide were tested in the MPTP mouse model of PD. RESULTS Analysing the receptor activating properties showed a balanced activation of GLP-1 and GIP receptors while not activating GLP-2 or glucagon receptors. DA-CH5 crossed the BBB better than other single or other dual receptor agonists. In a dose-response comparison, DA-CH5 was more effective than the GLP-1 receptor agonist exendin-4. When comparing the neuroprotective effect of DA-CH5 with Liraglutide, a GLP-1 analogue, both DA-CH5 and Liraglutide improved MPTP-induced motor impairments. In addition, the drugs reversed the decrease of the number of neurons expressing tyrosine hydroxylase (TH) in the SN, alleviated chronic inflammation, reduced lipid peroxidation, inhibited the apoptosis pathway (TUNEL assay) and increased autophagy -related proteins expression in the substantia nigra (SN) and striatum. Importantly, we found DA-CH5 was superior to Liraglutide in reducing microglia and astrocyte activation, improving mitochondrial activity by reducing the Bax/Bcl-2 ratio and normalising autophagy as found in abnormal expression of LC3 and p62. CONCLUSION The results demonstrate that the DA-CH5 is superior to liraglutide and could be a therapeutic treatment for PD.
Collapse
Affiliation(s)
- Lingyu Zhang
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Liping Zhang
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yanwei Li
- Department of Human Anatomy, Shaoyang Medical College, Shaoyang, Hunan, PR China
| | - Lin Li
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | | | - Mette Rosenkilde
- Department of Biomedical Science, University of Copenhagen, Copenhagen, Denmark
| | - Christian Hölscher
- Department of Second Hospital Neurology, Shanxi Medical University, Taiyuan, Shanxi, PR China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan province, PR China
| |
Collapse
|
45
|
Zhang J, Dong Y, Ju D, Feng J. Design, synthesis and biological evaluation of double fatty chain-modified glucagon-like peptide-1 conjugates. Bioorg Med Chem 2021; 44:116291. [PMID: 34216986 DOI: 10.1016/j.bmc.2021.116291] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/15/2021] [Accepted: 06/20/2021] [Indexed: 10/21/2022]
Abstract
Twelve double fatty chains and Aib8-Arg34-GLP-1 (7-37) were designed and obtained by microwave-assisted solid-phase synthesis. Then, twelve conjugates of Aib8-Arg34-GLP-1 (7-37) were synthesized in 1% triethylamine aqueous solution. Conjugates 2, 3, 6, 7, 10 and 11 showed better GLP-1 receptor activation potency than semaglutide. However, conjugates 2, 6 and 10 showed slightly worse glucose-lowering effects in vivo than semaglutide but better effects than conjugates 3, 7 and 11. The CD spectra of conjugates 2, 6 and 10 indicated that they had the same secondary structure as liraglutide and semaglutide. The receptor affinity results for conjugates 2, 6 and 10 measured by SPR (surface plasmon resonance) showed that conjugate 2 had higher receptor affinity than conjugates 6 and 10. In addition, albumin binding assays indicated that double fatty acid chains had obvious synergistic effects compared with single fatty acid chains. In conclusion, the structure-activity relationship of different side chains was summarized and one candidate, conjugate 2, was screened.
Collapse
Affiliation(s)
- Jinhua Zhang
- Department of Biological Medicines & Shanghai Engineering Research Centre of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China; China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Yuanzhen Dong
- China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Centre of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China.
| | - Jun Feng
- China State Institute of Pharmaceutical Industry, Shanghai, China; Shanghai Duomirui Biotechnology Ltd., Shanghai, China.
| |
Collapse
|
46
|
Ruan SD, Dong YZ, Lu JG, Zhao MJ, Lu WG, Feng J. Synthesis of a Novel PTH1–34 Analog with Increased Human Serum Albumin Affinity. PHARMACEUTICAL FRONTS 2021. [DOI: 10.1055/s-0041-1731299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Parathyroid hormone (PTH)1–34 is an effective peptide drug for osteoporosis therapy. However, the half-life of PTH1–34 in vivo is short, leading to the need for frequent injections of this drug during its treatment. To prolong the half-life of PTH1–34, a novel PTH1–34 analog was generated based on fatty acid generation, and its synthesis process included recombinant protein expression, side-chain modification, and peptide decoration. The PTH1–34 variant was expressed in Escherichia coli, with a single Lys (position 27) retained as a modification site. The side chain, –AEEA-γGlu-C18 diacid, was synthesized using 2-chlorotrityl chloride resin as a solid support, and then was conjugated to the PTH1-34 variant to form PTH-Lys27-AGC. Reversed-phase chromatography confirmed a high final purity (>98%) of the target compound; in vitro bioactivity tests showed that PTH-1 receptor potency of PTH-Lys27-AGC was comparable to that of the native PTH1–34. A competitive human serum albumin binding test demonstrated a high albumin affinity of PTH-Lys27-AGC in comparison to PTH1–34. In summary, we developed a novel PTH1–34 analog, PTH-Lys27-AGC, which may be a long-acting agent for osteoporosis treatment in the future.
Collapse
Affiliation(s)
- Si-Da Ruan
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Yuan-Zhen Dong
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Jian-Guang Lu
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Meng-Jia Zhao
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Wei-Gen Lu
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Jun Feng
- China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| |
Collapse
|
47
|
Micewicz ED, Damoiseaux RD, Deng G, Gomez A, Iwamoto KS, Jung ME, Nguyen C, Norris AJ, Ratikan JA, Ruchala P, Sayre JW, Schaue D, Whitelegge JP, McBride WH. Classes of Drugs that Mitigate Radiation Syndromes. Front Pharmacol 2021; 12:666776. [PMID: 34084139 PMCID: PMC8167044 DOI: 10.3389/fphar.2021.666776] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/27/2021] [Indexed: 11/13/2022] Open
Abstract
We previously reported several vignettes on types and classes of drugs able to mitigate acute and, in at least one case, late radiation syndromes in mice. Most of these had emerged from high throughput screening (HTS) of bioactive and chemical drug libraries using ionizing radiation-induced lymphocytic apoptosis as a readout. Here we report the full analysis of the HTS screen of libraries with 85,000 small molecule chemicals that identified 220 "hits." Most of these hits could be allocated by maximal common substructure analysis to one of 11 clusters each containing at least three active compounds. Further screening validated 23 compounds as being most active; 15 of these were cherry-picked based on drug availability and tested for their ability to mitigate acute hematopoietic radiation syndrome (H-ARS) in mice. Of these, five bore a 4-nitrophenylsulfonamide motif while 4 had a quinoline scaffold. All but two of the 15 significantly (p < 0.05) mitigated H-ARS in mice. We had previously reported that the lead 4-(nitrophenylsulfonyl)-4-phenylpiperazine compound (NPSP512), was active in mitigating multiple acute and late radiation syndromes in mice of more than one sex and strain. Unfortunately, the formulation of this drug had to be changed for regulatory reasons and we report here on the synthesis and testing of active analogs of NPSP512 (QS1 and 52A1) that have increased solubility in water and in vivo bioavailability while retaining mitigator activity against H-ARS (p < 0.0001) and other radiation syndromes. The lead quinoline 057 was also active in multiple murine models of radiation damage. Taken together, HTS of a total of 150,000 bioactive or chemical substances, combined with maximal common substructure analysis has resulted in the discovery of diverse groups of compounds that can mitigate H-ARS and at least some of which can mitigate multiple radiation syndromes when given starting 24 h after exposure. We discuss what is known about how these agents might work, and the importance of formulation and bioavailability.
Collapse
Affiliation(s)
- Ewa D. Micewicz
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| | - Robert D. Damoiseaux
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, CA, United States
- Department of Molecular and Medical Pharmacology, University of California at Los Angeles, Los Angeles, CA, United States
- Department of Bioengineering, Henry Samueli School of Engineering, University of California at Los Angeles, Los Angeles, CA, United States
| | - Gang Deng
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, CA, United States
| | - Adrian Gomez
- Pasarow Mass Spectrometry Laboratory, University of California at Los Angeles, Los Angeles, CA, United States
| | - Keisuke S. Iwamoto
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| | - Michael E. Jung
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, CA, United States
| | - Christine Nguyen
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| | | | - Josephine A. Ratikan
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| | - Piotr Ruchala
- Pasarow Mass Spectrometry Laboratory, University of California at Los Angeles, Los Angeles, CA, United States
| | - James W. Sayre
- Department of Biostatistics and Radiology, Fielding School of Public Health, University of California at Los Angeles, Los Angeles, CA, United States
| | - Dörthe Schaue
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| | - Julian P. Whitelegge
- Pasarow Mass Spectrometry Laboratory, University of California at Los Angeles, Los Angeles, CA, United States
| | - William H. McBride
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
48
|
Vana V, Lærke MK, Kleberg K, Mroz PA, Lindberg BL, Ekberg JH, Rehfeld JF, Schwartz TW, Hansen HS. Post-oral fat-induced satiation is mediated by endogenous CCK and GLP-1 in a fat self-administration mouse model. Physiol Behav 2021; 234:113315. [DOI: 10.1016/j.physbeh.2021.113315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022]
|
49
|
Sicinski K, Montanari V, Raman VS, Doyle JR, Harwood BN, Song YC, Fagan MP, Rios M, Haines DR, Kopin AS, Beinborn M, Kumar K. A Non-Perturbative Molecular Grafting Strategy for Stable and Potent Therapeutic Peptide Ligands. ACS CENTRAL SCIENCE 2021; 7:454-466. [PMID: 33791428 PMCID: PMC8006168 DOI: 10.1021/acscentsci.0c01237] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Indexed: 06/12/2023]
Abstract
The gut-derived incretin hormone, glucagon-like peptide-1 (GLP1), plays an important physiological role in attenuating post-prandial blood glucose excursions in part by amplifying pancreatic insulin secretion. Native GLP1 is rapidly degraded by the serine protease, dipeptidyl peptidase-4 (DPP4); however, enzyme-resistant analogues of this 30-amino-acid peptide provide an effective therapy for type 2 diabetes (T2D) and can curb obesity via complementary functions in the brain. In addition to its medical relevance, the incretin system provides a fertile arena for exploring how to better separate agonist function at cognate receptors versus susceptibility of peptides to DPP4-induced degradation. We have discovered that novel chemical decorations can make GLP1 and its analogues completely DPP4 resistant while fully preserving GLP1 receptor activity. This strategy is also applicable to other therapeutic ligands, namely, glucose-dependent insulinotropic polypeptide (GIP), glucagon, and glucagon-like peptide-2 (GLP2), targeting the secretin family of receptors. The versatility of the approach offers hundreds of active compounds based on any template that target these receptors. These observations should allow for rapid optimization of pharmacological properties and because the appendages are in a position crucial to receptor stimulation, they proffer the possibility of conferring "biased" signaling and in turn minimizing side effects.
Collapse
Affiliation(s)
- Kathleen
M. Sicinski
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Vittorio Montanari
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Venkata S. Raman
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Jamie R. Doyle
- Molecular
Cardiology Research Institute, Tufts Medical
Center, Boston, Massachusetts 02111, United States
| | - Benjamin N. Harwood
- Molecular
Cardiology Research Institute, Tufts Medical
Center, Boston, Massachusetts 02111, United States
| | - Yi Chi Song
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Micaella P. Fagan
- Department
of Neuroscience, Tufts University School
of Medicine, Boston, Massachusetts 02111, United States
| | - Maribel Rios
- Department
of Neuroscience, Tufts University School
of Medicine, Boston, Massachusetts 02111, United States
| | - David R. Haines
- Department of Chemistry, Wellesley College, Wellesley, Massachusetts 02481, United States
| | - Alan S. Kopin
- Molecular
Cardiology Research Institute, Tufts Medical
Center, Boston, Massachusetts 02111, United States
| | - Martin Beinborn
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
- Molecular
Cardiology Research Institute, Tufts Medical
Center, Boston, Massachusetts 02111, United States
| | - Krishna Kumar
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
50
|
Tinsley IC, Borner T, Swanson ML, Chepurny OG, Doebley SA, Kamat V, Sweet IR, Holz GG, Hayes MR, De Jonghe BC, Doyle RP. Synthesis, Optimization, and Biological Evaluation of Corrinated Conjugates of the GLP-1R Agonist Exendin-4. J Med Chem 2021; 64:3479-3492. [PMID: 33677970 PMCID: PMC8279408 DOI: 10.1021/acs.jmedchem.1c00185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
![]()
Corrination
is the conjugation of a corrin ring containing molecule,
such as vitamin B12 (B12) or B12 biosynthetic precursor
dicyanocobinamide (Cbi), to small molecules, peptides, or proteins
with the goal of modifying pharmacology. Recently, a corrinated GLP-1R
agonist (GLP-1RA) exendin-4 (Ex4) has been shown in vivo to have reduced penetration into the central nervous system relative
to Ex4 alone, producing a glucoregulatory GLP-1RA devoid of anorexia
and emesis. The study herein was designed to optimize the lead conjugate
for GLP-1R agonism and binding. Two specific conjugation sites were
introduced in Ex4, while also utilizing various linkers, so that it
was possible to identify Cbi conjugates of Ex4 that exhibit improved
binding and agonist activity at the GLP-1R. An optimized conjugate
(22), comparable with Ex4, was successfully screened
and subsequently assayed for insulin secretion in rat islets and in vivo in shrews for glucoregulatory and emetic behavior,
relative to Ex4.
Collapse
Affiliation(s)
- Ian C Tinsley
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
| | - Tito Borner
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania 19104, United States
| | - MacKenzie L Swanson
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
| | - Oleg G Chepurny
- Department of Medicine, State University of New York, Upstate Medical University, Syracuse, New York 13210, United States
| | - Sarah A Doebley
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania 19104, United States
| | - Varun Kamat
- Department of Medicine, University of Washington, Medicine Diabetes Institute, Seattle, Washington 98109, United States
| | - Ian R Sweet
- Department of Medicine, University of Washington, Medicine Diabetes Institute, Seattle, Washington 98109, United States
| | - George G Holz
- Department of Medicine, State University of New York, Upstate Medical University, Syracuse, New York 13210, United States
| | - Matthew R Hayes
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Bart C De Jonghe
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania 19104, United States
| | - Robert P Doyle
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States.,Department of Medicine, State University of New York, Upstate Medical University, Syracuse, New York 13210, United States
| |
Collapse
|