1
|
Jiao Q, Huang Y, He J, Xu Y. Advances in Oral Biomacromolecule Therapies for Metabolic Diseases. Pharmaceutics 2025; 17:238. [PMID: 40006605 PMCID: PMC11859201 DOI: 10.3390/pharmaceutics17020238] [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: 01/18/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic diseases like obesity and diabetes are on the rise, and therapies with biomacromolecules (such as proteins, peptides, antibodies, and oligonucleotides) play a crucial role in their treatment. However, these drugs are traditionally injected. For patients with chronic diseases (e.g., metabolic diseases), long-term injections are accompanied by inconvenience and low compliance. Oral administration is preferred, but the delivery of biomacromolecules is challenging due to gastrointestinal barriers. In this article, we introduce the available biomacromolecule drugs for the treatment of metabolic diseases. The gastrointestinal barriers to oral drug delivery and strategies to overcome these barriers are also explored. We then discuss strategies for alleviating metabolic defects, including glucose metabolism, lipid metabolism, and energy metabolism, with oral biomacromolecules such as insulin, glucagon-like peptide-1 receptor agonists, proprotein convertase subtilisin/kexin type 9 inhibitors, fibroblast growth factor 21 analogues, and peptide YY analogues.
Collapse
Affiliation(s)
- Qiuxia Jiao
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Huang
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yining Xu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
2
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [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: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
3
|
Camilleri M, Acosta A. Newer pharmacological interventions directed at gut hormones for obesity. Br J Pharmacol 2024; 181:1153-1164. [PMID: 37917871 PMCID: PMC10947960 DOI: 10.1111/bph.16278] [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: 06/27/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
The objective is to review the newer pharmacological interventions for obesity, specifically single, dual and triple incretin receptor agonists that are either available or in the pipeline for treatment of obesity. The three incretin receptor targets are glucagon like peptide-1 (GLP-1), glucose-dependent insulinotropic peptide (GIP) and glucagon. There are several approved single or dual incretin agonists which can be administered subcutaneously daily (e.g., liraglutide) or weekly (e.g., semaglutide, dulaglutide, and exenatide QW), and other experimental dual or triple incretin agonists. Analogues of amylin, peptide YY and oxyntomodulin, as well as the combination of a GLP1R agonist and GIPR antagonist also are in development. Oral semaglutide (administered daily) is approved for type 2 diabetes mellitus and is on track for regulatory review for obesity. The review includes specifically perspectives on the effects of these mechanisms and pharmacological agents on gastric emptying, which contribute to satiation and weight loss, in addition to the established evidence on effects on central mechanisms controlling appetite. In the future, it is anticipated that small molecule GLP-1 receptor agonists (e.g., oral danuglipron) will be developed for treating obesity. These pharmacological agents are having significant impact on glycaemic control and obesity and on their co-morbidities.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Mayo Clinic, Rochester, Minnesota, USA
| | - Andres Acosta
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
4
|
Solis-Herrera C, Kane MP, Triplitt C. Current Understanding of Sodium N-(8-[2-Hydroxylbenzoyl] Amino) Caprylate (SNAC) as an Absorption Enhancer: The Oral Semaglutide Experience. Clin Diabetes 2023; 42:74-86. [PMID: 38230324 PMCID: PMC10788673 DOI: 10.2337/cd22-0118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Oral administration of peptide therapeutics faces challenges because of the distinct environment of the gastrointestinal tract. An oral formulation of semaglutide, a glucagon-like peptide 1 receptor agonist, was approved by the U.S. Food and Drug Administration in 2019 as a peptide therapy for the treatment of type 2 diabetes. Oral semaglutide uses sodium N-(8-[2-hydroxybenzoyl] amino) caprylate (SNAC) technology to enhance the absorption of semaglutide in the stomach and protect it from degradation by gastric enzymes. This article presents a summary of studies investigating SNAC technology as an absorption enhancer for a number of molecules and, in particular, explores how SNAC, once coformulated with oral semaglutide, facilitates increased absorption and bioavailability. Practical advice and dispensing information for pharmacists is also provided.
Collapse
Affiliation(s)
- Carolina Solis-Herrera
- Division of Diabetes, University of Texas Health Science Center, San Antonio, TX
- Division of Endocrinology, University of Texas Health Science Center, San Antonio, TX
| | - Michael P. Kane
- Department of Pharmacy Practice, Albany College of Pharmacy and Health Sciences, Albany, NY
| | - Curtis Triplitt
- Division of Diabetes, University of Texas Health Science Center, San Antonio, TX
| |
Collapse
|
5
|
Gastrointestinal Permeation Enhancers for the Development of Oral Peptide Pharmaceuticals. Pharmaceuticals (Basel) 2022; 15:ph15121585. [PMID: 36559036 PMCID: PMC9781085 DOI: 10.3390/ph15121585] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Recently, two oral-administered peptide pharmaceuticals, semaglutide and octreotide, have been developed and are considered as a breakthrough in peptide and protein drug delivery system development. In 2019, the Food and Drug Administration (FDA) approved an oral dosage form of semaglutide developed by Novo Nordisk (Rybelsus®) for the treatment of type 2 diabetes. Subsequently, the octreotide capsule (Mycapssa®), developed through Chiasma's Transient Permeation Enhancer (TPE) technology, also received FDA approval in 2020 for the treatment of acromegaly. These two oral peptide products have been a significant success; however, a major obstacle to their oral delivery remains the poor permeability of peptides through the intestinal epithelium. Therefore, gastrointestinal permeation enhancers are of great relevance for the development of subsequent oral peptide products. Sodium salcaprozate (SNAC) and sodium caprylate (C8) have been used as gastrointestinal permeation enhancers for semaglutide and octreotide, respectively. Herein, we briefly review two approved products, Rybelsus® and Mycapssa®, and discuss the permeation properties of SNAC and medium chain fatty acids, sodium caprate (C10) and C8, focusing on Eligen technology using SNAC, TPE technology using C8, and gastrointestinal permeation enhancement technology (GIPET) using C10.
Collapse
|
6
|
Wang Y, Wu Y, Wang A, Wang A, Alkhalidy H, Helm R, Zhang S, Ma H, Zhang Y, Gilbert E, Xu B, Liu D. An olive-derived elenolic acid stimulates hormone release from L-cells and exerts potent beneficial metabolic effects in obese diabetic mice. Front Nutr 2022; 9:1051452. [PMID: 36386896 PMCID: PMC9664001 DOI: 10.3389/fnut.2022.1051452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/11/2022] [Indexed: 06/16/2023] Open
Abstract
Insulin resistance and progressive decline in functional β-cell mass are two key factors for developing type 2 diabetes (T2D), which is largely driven by overweight and obesity, a significant obstacle for effective metabolic control in many patients with T2D. Thus, agents that simultaneously ameliorate obesity and act on multiple pathophysiological components could be more effective for treating T2D. Here, we report that elenolic acid (EA), a phytochemical, is such a dual-action agent. we show that EA dose-dependently stimulates GLP-1 secretion in mouse clonal L-cells and isolated mouse ileum crypts. In addition, EA induces L-cells to secrete peptide YY (PYY). EA induces a rapid increase in intracellular [Ca2+]i and the production of inositol trisphosphate in L-cells, indicating that EA activates phospholipase C (PLC)-mediated signaling. Consistently, inhibition of (PLC) or Gαq ablates EA-stimulated increase of [Ca2+]i and GLP-1 secretion. In vivo, a single dose of EA acutely stimulates GLP-1 and PYY secretion in mice, accompanied with an improved glucose tolerance and insulin levels. Oral administration of EA at a dose of 50 mg/kg/day for 2 weeks normalized the fasting blood glucose and restored glucose tolerance in high-fat diet-induced obese (DIO) mice to levels that were comparable to chow-fed mice. In addition, EA suppresses appetite, reduces food intake, promotes weight loss, and reverses perturbated metabolic variables in obese mice. These results suggest that EA could be a dual-action agent as an alternative or adjuvant treatment for both T2D and obesity.
Collapse
Affiliation(s)
- Yao Wang
- Department of Human Nutrition, Foods, and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Yajun Wu
- Department of Human Nutrition, Foods, and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Aiping Wang
- College of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Aihua Wang
- Department of Biochemistry, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Hana Alkhalidy
- Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid, Jordan
| | - Richard Helm
- Department of Biochemistry, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Shijun Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States
| | - Hongguang Ma
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States
| | - Elizabeth Gilbert
- School of Animal Sciences, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Bin Xu
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, NC, United States
| | - Dongmin Liu
- Department of Human Nutrition, Foods, and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA, United States
- Virginia Tech Drug Discovery Center, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
7
|
Wortha SM, Wüsten KA, Witte VA, Bössel N, Keßler W, Vogelgesang A, Flöel A. Gastrointestinal Hormones in Healthy Adults: Reliability of Repeated Assessments and Interrelations with Eating Habits and Physical Activity. Nutrients 2021; 13:nu13113809. [PMID: 34836065 PMCID: PMC8624073 DOI: 10.3390/nu13113809] [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: 09/18/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/04/2022] Open
Abstract
Background: Gastrointestinal hormones (GIHs) are crucial for the regulation of a variety of physiological functions and have been linked to hunger, satiety, and appetite control. Thus, they might constitute meaningful biomarkers in longitudinal and interventional studies on eating behavior and body weight control. However, little is known about the physiological levels of GIHs, their intra-individual stability over time, and their interaction with other metabolic and lifestyle-related parameters. Therefore, the aim of this pilot study is to investigate the intra-individual stability of GIHs in normal-weight adults over time. Methods: Plasma concentrations of ghrelin, leptin, GLP-1 (glucagon-like-peptide), and PP (pancreatic polypeptide) were assessed by enzyme-linked immunosorbent assay (ELISA) in 17 normal-weight, healthy adults in a longitudinal design at baseline and at follow-up six months later. The reliability of the measurements was estimated using intra-class correlation (ICC). In a second step, we considered the stability of GIH levels after controlling for changes in blood glucose and hemoglobin A1 (HbA1c) as well as self-reported physical activity and dietary habits. Results: We found excellent reliability for ghrelin, good reliability for GLP1 and PP, and moderate reliability for leptin. After considering glucose, HbA1c, physical activity, and dietary habits as co-variates, the reliability of ghrelin, GLP1, and PP did not change significantly; the reliability of leptin changed to poor reliability. Conclusions: The GIHs ghrelin, GLP1, and PP demonstrated good to excellent test–retest reliability in healthy individuals, a finding that was not modified after adjusting for glucose control, physical activity, or dietary habits. Leptin showed only moderate to poor reliability, which might be linked to weight fluctuations, albeit small, between baseline and follow-up assessment in our study sample. Together, these findings support that ghrelin, GLP1, and PP might be further examined as biomarkers in studies on weight control, with GLP1 and PP serving as anorexic markers and ghrelin as an orexigenic marker. Additional reliability studies in obese individuals are necessary to verify or refute our findings for this cohort.
Collapse
Affiliation(s)
- Silke M. Wortha
- Department of Neurology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.M.W.); (K.A.W.); (N.B.); (A.V.)
| | - Katharina A. Wüsten
- Department of Neurology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.M.W.); (K.A.W.); (N.B.); (A.V.)
| | - Veronica A. Witte
- Department of Cognitive Neurology, University of Leipzig Medical Center, 04103 Leipzig, Germany;
- Max Planck Institute for Cognitive and Brain Sciences, 04103 Leipzig, Germany
| | - Nicole Bössel
- Department of Neurology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.M.W.); (K.A.W.); (N.B.); (A.V.)
| | - Wolfram Keßler
- Department of General, Visceral, Thoracic and Vascular Surgery, University Medicine, 17475 Greifswald, Germany;
| | - Antje Vogelgesang
- Department of Neurology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.M.W.); (K.A.W.); (N.B.); (A.V.)
| | - Agnes Flöel
- Department of Neurology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.M.W.); (K.A.W.); (N.B.); (A.V.)
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17489 Greifswald, Germany
- Correspondence:
| |
Collapse
|
8
|
Alhabeeb H, AlFaiz A, Kutbi E, AlShahrani D, Alsuhail A, AlRajhi S, Alotaibi N, Alotaibi K, AlAmri S, Alghamdi S, AlJohani N. Gut Hormones in Health and Obesity: The Upcoming Role of Short Chain Fatty Acids. Nutrients 2021; 13:nu13020481. [PMID: 33572661 PMCID: PMC7911102 DOI: 10.3390/nu13020481] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 12/21/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
We are currently facing an obesity pandemic, with worldwide obesity rates having tripled since 1975. Obesity is one of the main risk factors for the development of non-communicable diseases, which are now the leading cause of death worldwide. This calls for urgent action towards understanding the underlying mechanisms behind the development of obesity as well as developing more effective treatments and interventions. Appetite is carefully regulated in humans via the interaction between the central nervous system and peripheral hormones. This involves a delicate balance in external stimuli, circulating satiating and appetite stimulating hormones, and correct functioning of neuronal signals. Any changes in this equilibrium can lead to an imbalance in energy intake versus expenditure, which often leads to overeating, and potentially weight gain resulting in overweight or obesity. Several lines of research have shown imbalances in gut hormones are found in those who are overweight or obese, which may be contributing to their condition. Therefore, this review examines the evidence for targeting gut hormones in the treatment of obesity by discussing how their dysregulation influences food intake, the potential possibility of altering the circulating levels of these hormones for treating obesity, as well as the role of short chain fatty acids and protein as novel treatments.
Collapse
Affiliation(s)
- Habeeb Alhabeeb
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
- Correspondence:
| | - Ali AlFaiz
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Emad Kutbi
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Dayel AlShahrani
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Abdullah Alsuhail
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Saleh AlRajhi
- Family Medicine, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia;
| | - Nemer Alotaibi
- College of Medicine, Shaqra University, Shaqra 11961, Saudi Arabia; (N.A.); (K.A.)
| | - Khalid Alotaibi
- College of Medicine, Shaqra University, Shaqra 11961, Saudi Arabia; (N.A.); (K.A.)
| | - Saad AlAmri
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Saleh Alghamdi
- Research Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia; (A.A.); (E.K.); (D.A.); (A.A.); (S.A.); (S.A.)
| | - Naji AlJohani
- Obesity, Endocrine, and Metabolism Center, King Fahad Medical City—KFMC, Riyadh 11525, Saudi Arabia;
| |
Collapse
|
9
|
Food Addiction and Tobacco Use Disorder: Common Liability and Shared Mechanisms. Nutrients 2020; 12:nu12123834. [PMID: 33334010 PMCID: PMC7765398 DOI: 10.3390/nu12123834] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
As food addiction is being more commonly recognized within the scientific community, parallels can be drawn between it and other addictive substance use disorders, including tobacco use disorder. Given that both unhealthy diets and smoking are leading risk factors for disability and death, a greater understanding of how food addiction and tobacco use disorder overlap with one another is necessary. This narrative review aimed to highlight literature that investigated prevalence, biology, psychology, and treatment options of food addiction and tobacco use disorder. Published studies up to August 2020 and written in English were included. Using a biopsychosocial lens, each disorder was assessed together and separately, as there is emerging evidence that the two disorders can develop concurrently or sequentially within individuals. Commonalities include but are not limited to the dopaminergic neurocircuitry, gut microbiota, childhood adversity, and attachment insecurity. In addition, the authors conducted a feasibility study with the purpose of examining the association between food addiction symptoms and tobacco use disorder among individuals seeking tobacco use disorder treatment. To inform future treatment approaches, more research is necessary to identify and understand the overlap between the two disorders.
Collapse
|
10
|
Maher S, Geoghegan C, Brayden DJ. Intestinal permeation enhancers to improve oral bioavailability of macromolecules: reasons for low efficacy in humans. Expert Opin Drug Deliv 2020; 18:273-300. [PMID: 32937089 DOI: 10.1080/17425247.2021.1825375] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Intestinal permeation enhancers (PEs) are substances that transiently alter the intestinal epithelial barrier to facilitate permeation of macromolecules with low oral bioavailability (BA). While a number of PEs have progressed to clinical testing in conventional formulations with macromolecules, there has been only low single digit increases in oral BA, irrespective of whether the drug met primary or secondary clinical endpoints. AREAS COVERED This article considers the causes of sub-optimal BA of macromolecules from PE dosage forms and suggests approaches that may improve performance in humans. EXPERT OPINION Permeation enhancement is most effective when the PE is co-localized with the macromolecule at the epithelial surface. Conditions in the GI tract impede optimal co-localization. Novel delivery systems that limit dilution and spreading of the PE and macromolecule in the small intestine have attempted to replicate promising enhancement efficacy observed in static drug delivery models.
Collapse
Affiliation(s)
- Sam Maher
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Caroline Geoghegan
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
11
|
McVeay C, Steinert RE, Fitzgerald PCE, Ullrich SS, Horowitz M, Feinle-Bisset C. Effects of intraduodenal coadministration of lauric acid and leucine on gut motility, plasma cholecystokinin, and energy intake in healthy men. Am J Physiol Regul Integr Comp Physiol 2020; 318:R790-R798. [PMID: 32160019 DOI: 10.1152/ajpregu.00352.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The fatty acid, lauric acid (C12), and the amino acid, leucine (Leu) stimulate gut hormones, including CCK, associated with suppression of energy intake. In our recent study, intraduodenal infusion of a combination of C12 and l-tryptophan, at loads that individually did not affect energy intake, reduced energy intake substantially, associated with much greater stimulation of CCK. We have now investigated whether combined administration of C12 and Leu would enhance the intake-suppressant effects of each nutrient, when given at loads that each suppress energy intake individually. Sixteen healthy, lean males (age: 23 ± 2 yr) received, in randomized, double-blind fashion, 90-min intraduodenal infusions of control (saline), C12 (0.4 kcal/min), Leu (0.45 kcal/min), or C12+Leu (0.85 kcal/min). Antropyloroduodenal pressures were measured continuously and plasma CCK at 15-min intervals, and energy intake from a standardized buffet-meal, consumed immediately postinfusion, was quantified. All nutrient infusions stimulated plasma CCK compared with control (P < 0.05). Moreover, C12 and C12+Leu stimulated CCK compared with Leu (P < 0.05) (mean concentration, pmol/L; control: 2.3 ± 0.3, C12: 3.8 ± 0.3, Leu: 2.7 ± 0.3, and C12+Leu: 4.0 ± 0.4). C12+Leu, but not C12 or Leu, stimulated pyloric pressures (P < 0.05). C12+Leu and C12 reduced energy intake (P < 0.05), and there was a trend for Leu to reduce (P = 0.06) energy intake compared with control, with no differences between the three nutrient treatments (kcal; control: 1398 ± 84, C12: 1226 ± 80, Leu: 1260 ± 92, and C12+Leu: 1208 ± 83). In conclusion, combination of C12 and Leu, at the loads given, did not reduce energy intake beyond their individual effects, possibly because maximal effects had been evoked.
Collapse
Affiliation(s)
- Christina McVeay
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Robert E Steinert
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Penelope C E Fitzgerald
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Sina S Ullrich
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Michael Horowitz
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Christine Feinle-Bisset
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| |
Collapse
|
12
|
Kokkinos A, Tsilingiris D, le Roux CW, Rubino F, Mantzoros CS. Will medications that mimic gut hormones or target their receptors eventually replace bariatric surgery? Metabolism 2019; 100:153960. [PMID: 31412266 DOI: 10.1016/j.metabol.2019.153960] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 02/07/2023]
Abstract
Bariatric surgery is currently the most effective therapeutic modality through which sustained beneficial effects on weight loss and metabolic improvement are achieved. During recent years, indications for bariatric surgery have been expanded to include cases of poorly controlled type 2 (T2DM) diabetes mellitus in lesser extremes of body weight. A spectrum of the beneficial effects of surgery is attributed to robust changes of postprandial gut peptide responses that are observed post operatively. Consolidated knowledge regarding gut peptide physiology as well as emerging new evidence shedding light on the mode of action of previously overlooked gut hormones provide appealing potential obesity and T2DM therapeutic perspectives. The accumulation of evidence from the effect of exogenous administration of native gut peptides alone or in combinations to humans as well as the development of mimetic agents exerting agonistic effects on combinations of gut hormone receptors pave the way for future integrated gut peptide-based treatments, which may mimic the effects of bariatric surgery.
Collapse
Affiliation(s)
- Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece.
| | - Dimitrios Tsilingiris
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Carel W le Roux
- Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - Francesco Rubino
- Department of Metabolic and Bariatric Surgery, Diabetes and Nutritional Science Division, King's College Hospital, London, United Kingdom
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| |
Collapse
|
13
|
Jones ES, Nunn N, Chambers AP, Østergaard S, Wulff BS, Luckman SM. Modified Peptide YY Molecule Attenuates the Activity of NPY/AgRP Neurons and Reduces Food Intake in Male Mice. Endocrinology 2019; 160:2737-2747. [PMID: 31074796 PMCID: PMC6806261 DOI: 10.1210/en.2019-00100] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/06/2019] [Indexed: 01/15/2023]
Abstract
To study the effects of an analog of the gut-produced hormone peptide YY (PYY3-36), which has increased selectivity for the Y2 receptor; specifically, to record its effects on food intake and on hypothalamic neuropeptide Y/agouti-related peptide (NPY/AgRP) neuron activity. NNC0165-1273, a modified form of the peptide hormone PYY3-36 with potent selectivity at Y2 receptor (>5000-fold over Y1, 1250-fold over Y4, and 650-fold over Y5 receptor), was tested in vivo and in vitro in mouse models. NNC0165-1273 has fivefold lower relative affinity for Y2 compared with PYY3-36, but >250-, 192-, and 400-fold higher selectivity, respectively, for the Y1, Y4, and Y5 receptors. NNC0165-1273 produced a reduction in nighttime feeding at a dose at which PYY3-36 loses efficacy. The normal behavioral satiety sequence observed suggests that NNC0165-1273 is not nauseating and, instead, reduces food intake by producing early satiety. Additionally, NNC0165-1273 blocked ghrelin-induced cFos expression in NPY/AgRP neurons. In vitro electrophysiological recordings showed that, opposite to ghrelin, NNC0165-1273 hyperpolarized NPY/AgRP neurons and reduced action potential frequency. Administration of NNC0165-1273 via subcutaneous osmotic minipump caused a dose-dependent decrease in body weight and fat mass in an obese mouse model. Finally, NNC0165-1273 attenuated the feeding response when NPY/AgRP neurons were activated using ghrelin or more selectively with designer receptors. NNC0165-1273 is nonnauseating and stimulates a satiety response through, at least in part, a direct action on hypothalamic NPY/AgRP neurons. Modification of PYY3-36 to produce compounds with increased affinity to Y2 receptors may be useful as antiobesity therapies in humans.
Collapse
Affiliation(s)
- Edward S Jones
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Nicolas Nunn
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Adam P Chambers
- GLP-1 & T2D Pharmacology, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Søren Østergaard
- Research Chemistry 2, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Birgitte S Wulff
- Obesity Research, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Simon M Luckman
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
14
|
Rose F, Bloom S, Tan T. Novel approaches to anti-obesity drug discovery with gut hormones over the past 10 years. Expert Opin Drug Discov 2019; 14:1151-1159. [DOI: 10.1080/17460441.2019.1646243] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Frances Rose
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Stephen Bloom
- Department of Investigative Medicine, Imperial College London, London, UK
| | - Tricia Tan
- Department of Investigative Medicine, Imperial College London, London, UK
| |
Collapse
|
15
|
Shao J, Chen MJ, Kuehl PJ, Hochhaus G. Pharmacokinetic and pharmacodynamic modeling of gut hormone peptide YY (3-36) after pulmonary delivery. Drug Dev Ind Pharm 2019; 45:1101-1110. [PMID: 31039626 DOI: 10.1080/03639045.2019.1593443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Peptide YY(3-36) (PYY(3-36)) is an endogenous appetite suppressing peptide. The present research was to perform pharmacokinetic/pharmacodynamic (PK/PD) analysis for predicting the concentration- and response-time profiles of PYY(3-36) after systemic and pulmonary delivery in mice, with the goal of suggesting a potential pulmonary dosing regimen in humans. A PK/PD model was developed to describe PYY(3-36) plasma concentration - and relative food intake rate ratio (as % of control) - time profiles after intraperitoneal and subcutaneous administration, and inhalation in mice. The absorption of inhaled PYY(3-36) from the lungs of mice could only be described with a combined slow (absorption rate of 0.147 L/h) and fast (absorption rate of 104.4 L/h) absorption process, presumably related to absorption from the central and peripheral regions of the lungs. The estimates for IC50 and Imax were 6.8 ng/mL and 63.5%, respectively, based on inhibitory Emax model. The PK parameters, such as clearance (CL), volume of distribution at steady state (Vdss), and the absorption rates (ka), were then scaled to human's. The scaled human CL and Vdss for obese subjects were 24.8 L/h and 9.0 L, respectively. The model predicted human plasma PYY(3-36) concentrations agreed reasonably well with placebo-normalized plasma PYY(3-36) concentrations after short-term infusion and SC injection in literature. An inhalation dose of PYY(3-36) of about 100 µg was proposed for obese subjects based on simulations. This PK/PD analysis satisfactorily described PYY(3-36) concentration-time and relative food intake rate ratio- time profiles at all doses and routes. The developed model might facilitate the inhalation dose selection of PYY(3-36).
Collapse
Affiliation(s)
- Jie Shao
- a Department of Pharmaceutics, College of Pharmacy , University of Florida , Gainesville , Florida , USA
| | - Mong-Jen Chen
- a Department of Pharmaceutics, College of Pharmacy , University of Florida , Gainesville , Florida , USA
| | - Philip J Kuehl
- b Lovelace Respiratory Research Institute , Albuquerque , New Mexico , USA
| | - Guenther Hochhaus
- a Department of Pharmaceutics, College of Pharmacy , University of Florida , Gainesville , Florida , USA
| |
Collapse
|
16
|
McVeay C, Fitzgerald PCE, Ullrich SS, Steinert RE, Horowitz M, Feinle-Bisset C. Effects of intraduodenal administration of lauric acid and L-tryptophan, alone and combined, on gut hormones, pyloric pressures, and energy intake in healthy men. Am J Clin Nutr 2019; 109:1335-1343. [PMID: 31051504 DOI: 10.1093/ajcn/nqz020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/25/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The fatty acid, lauric acid ('C12'), and the amino acid, L-tryptophan ('Trp'), modulate gastrointestinal functions including gut hormones and pyloric pressures, which are important for the regulation of energy intake, and both potently suppress energy intake. OBJECTIVE We hypothesized that the intraduodenal administration of C12 and Trp, at loads that do not affect energy intake individually, when combined will reduce energy intake, which is associated with greater modulation of gut hormones and pyloric pressures. DESIGN Sixteen healthy, lean males (age: 24 ± 1.5 y) received 90-min intraduodenal infusions of saline (control), C12 (0.3 kcal/min), Trp (0.1 kcal/min), or C12 + Trp (0.4 kcal/min), in a randomized, double-blind, cross-over study. Antropyloroduodenal pressures were measured continuously, and plasma cholecystokinin (CCK), ghrelin, and glucagon-like peptide-1 (GLP-1) concentrations, appetite perceptions, and gastrointestinal symptoms at 15-min intervals. Immediately after the infusions, energy intake from a standardized buffet meal was quantified. RESULTS C12 + Trp markedly reduced energy intake (kcal; control: 1,232 ± 72, C12: 1,180 ± 82, Trp: 1,269 ± 73, C12 + Trp: 1,056 ± 106), stimulated plasma CCK (AUC(area under the curve)0-90 min, pmol/L*min; control: 21 ± 8; C12: 129 ± 15; Trp: 97 ± 16; C12 + Trp: 229 ± 22) and GLP-1 (AUC0-90 min, pmol/L*min; control: 102 ± 41; C12: 522 ± 102; Trp: 198 ± 63; C12 + Trp: 545 ± 138), and suppressed ghrelin (AUC0-90 min, pg/mL*min; control: -3,433 ± 2,647; C12: -11,825 ± 3,521; Trp: -8,417 ± 3,734; C12 + Trp: -18,188 ± 4,165) concentrations, but did not stimulate tonic, or phasic, pyloric pressures, compared with the control (all P < 0.05), or have adverse effects. C12 and Trp each stimulated CCK (P < 0.05), but to a lesser degree than C12 + Trp, and did not suppress energy intake or ghrelin. C12, but not Trp, stimulated GLP-1 (P < 0.05) and phasic pyloric pressures (P < 0.05), compared with the control. CONCLUSION The combined intraduodenal administration of C12 and Trp, at loads that individually do not affect energy intake, substantially reduces energy intake, which is associated with a marked stimulation of CCK and suppression of ghrelin. The study was registered as a clinical trial at the Australian and New Zealand Clinical Trial Registry (www.anzctr.org.au,) as 12613000899741.
Collapse
Affiliation(s)
- Christina McVeay
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Penelope C E Fitzgerald
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Sina S Ullrich
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Robert E Steinert
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Michael Horowitz
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Christine Feinle-Bisset
- Adelaide Medical School and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| |
Collapse
|
17
|
|
18
|
Pilitsi E, Farr OM, Polyzos SA, Perakakis N, Nolen-Doerr E, Papathanasiou AE, Mantzoros CS. Pharmacotherapy of obesity: Available medications and drugs under investigation. Metabolism 2019; 92:170-192. [PMID: 30391259 DOI: 10.1016/j.metabol.2018.10.010] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/13/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
Obesity is a chronic disease with a continuously rising prevalence that currently affects more than half a billion people worldwide. Energy balance and appetite are highly regulated via central and peripheral mechanisms, and weight loss triggers a homeostatic response leading to weight regain. Lifestyle and behavioral modifications are the cornerstones of obesity management; however, they often fail to achieve or sustain long-term weight loss. Pharmacotherapy added onto lifestyle modifications results in an additional, albeit limited, weight reduction. Regardless, this weight reduction of 5-10% conveys multiple cardiovascular and metabolic benefits. In this review, evidence on the food and drug administration (FDA)-approved medications, i.e., orlistat, lorcaserin, phentermine/topiramate, liraglutide and naltrexone/bupropion, is summarized. Furthermore, anti-obesity agents in the pipeline for potential future therapeutic use are presented.
Collapse
Affiliation(s)
- Eleni Pilitsi
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215
| | - Olivia M Farr
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215.
| | - Stergios A Polyzos
- First Department of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Perakakis
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215
| | - Eric Nolen-Doerr
- Department of Medicine, Boston Medical Center, Boston, MA, 02118, United States of America
| | - Aimilia-Eirini Papathanasiou
- Division of Pediatric Newborn Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA 02215, United States of America
| | - Christos S Mantzoros
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215; Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Brandt SJ, Müller TD, DiMarchi RD, Tschöp MH, Stemmer K. Peptide-based multi-agonists: a new paradigm in metabolic pharmacology. J Intern Med 2018; 284:581-602. [PMID: 30230640 DOI: 10.1111/joim.12837] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Obesity and its comorbidities, such as type 2 diabetes, are pressing worldwide health concerns. Available anti-obesity treatments include weight loss pharmacotherapies and bariatric surgery. Whilst surgical interventions typically result in significant and sustained weight loss, available pharmacotherapies are far less effective, typically decreasing body weight by no more than 5-10%. An emerging class of multi-agonist drugs may eventually bridge this gap. This new class of specially tailored drugs hybridizes the amino acid sequences of key metabolic hormones into one single entity with enhanced potency and sustained action. Successful examples of this strategy include multi-agonist drugs targeting the receptors for glucagon-like peptide-1 (GLP-1), glucagon and the glucose-dependent insulinotropic polypeptide (GIP). Due to the simultaneous activity at several metabolically relevant receptors, these multi-agonists offer improved body weight loss and glucose tolerance relative to their constituent monotherapies. Further advancing this concept, chimeras were generated that covalently link nuclear acting hormones such as oestrogen, thyroid hormone (T3 ) or dexamethasone to peptide hormones such as GLP-1 or glucagon. The benefit of this strategy is to restrict the nuclear hormone action exclusively to cells expressing the peptide hormone receptor, thereby maximizing combinatorial metabolic efficacy of both drug constituents in the target cells whilst preventing the nuclear hormone cargo from entering and acting on cells devoid of the peptide hormone receptor, in which the nuclear hormone might have unwanted effects. Many of these multi-agonists are in preclinical and clinical development and may represent new and effective tools in the fight against obesity and its comorbidities.
Collapse
Affiliation(s)
- S J Brandt
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - T D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - R D DiMarchi
- Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - M H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - K Stemmer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
20
|
Tyagi P, Pechenov S, Anand Subramony J. Oral peptide delivery: Translational challenges due to physiological effects. J Control Release 2018; 287:167-176. [DOI: 10.1016/j.jconrel.2018.08.032] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 01/15/2023]
|
21
|
Hope DCD, Tan TMM, Bloom SR. No Guts, No Loss: Toward the Ideal Treatment for Obesity in the Twenty-First Century. Front Endocrinol (Lausanne) 2018; 9:442. [PMID: 30158899 PMCID: PMC6104129 DOI: 10.3389/fendo.2018.00442] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 07/17/2018] [Indexed: 12/25/2022] Open
Abstract
Over the last century, our knowledge of the processes which control appetite and weight regulation has developed significantly. The understanding of where gut hormones fit into the control of energy homeostasis in addition to the rapid advancement of pharmacotherapeutics has paved the way for the development of novel gut hormone analogs to target weight loss. Currently, bariatric surgery remains the most efficacious treatment for obesity. The emergence of gut hormone analogs may provide a useful non-surgical addition to the armamentarium in treating obesity. Simply targeting single gut hormone pathways may be insufficiently efficacious, and combination/multiple-agonist approaches may be necessary to obtain the results required for clear clinical impact.
Collapse
Affiliation(s)
- David C D Hope
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, United Kingdom
| | - Tricia M M Tan
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, United Kingdom
| | - Stephen R Bloom
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, United Kingdom
| |
Collapse
|
22
|
Chepurny OG, Bonaccorso RL, Leech CA, Wöllert T, Langford GM, Schwede F, Roth CL, Doyle RP, Holz GG. Chimeric peptide EP45 as a dual agonist at GLP-1 and NPY2R receptors. Sci Rep 2018; 8:3749. [PMID: 29491394 PMCID: PMC5830615 DOI: 10.1038/s41598-018-22106-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/16/2018] [Indexed: 02/07/2023] Open
Abstract
We report the design and target validation of chimeric peptide EP45, a novel 45 amino acid monomeric dual agonist peptide that contains amino acid sequence motifs present within the blood glucose-lowering agent exendin-4 (Ex-4) and the appetite-suppressing agent PYY(3-36). In a new high-throughput FRET assay that provides real-time kinetic information concerning levels of cAMP in living cells, EP45 recapitulates the action of Ex-4 to stimulate cAMP production via the glucagon-like peptide-1 receptor (GLP-1R), while also recapitulating the action of PYY(3-36) to inhibit cAMP production via the neuropeptide Y2 receptor (NPY2R). EP45 fails to activate glucagon or GIP receptors, whereas for cells that co-express NPY2R and adenosine A2B receptors, EP45 acts in an NPY2R-mediated manner to suppress stimulatory effects of adenosine on cAMP production. Collectively, such findings are remarkable in that they suggest a new strategy in which the co-existing metabolic disorders of type 2 diabetes and obesity will be treatable using a single peptide such as EP45 that lowers levels of blood glucose by virtue of its GLP-1R-mediated effect, while simultaneously suppressing appetite by virtue of its NPY2R-mediated effect.
Collapse
Affiliation(s)
- Oleg G Chepurny
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, 505 Irving Avenue, Syracuse, NY, 13210, USA
| | - Ron L Bonaccorso
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - Colin A Leech
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, 505 Irving Avenue, Syracuse, NY, 13210, USA
| | - Torsten Wöllert
- Department of Biology, Syracuse University, Syracuse, NY, 13244, USA
| | - George M Langford
- Department of Biology, Syracuse University, Syracuse, NY, 13244, USA
| | - Frank Schwede
- BIOLOG Life Science Institute, 28199, Bremen, Germany
| | - Christian L Roth
- Center for Integrative Brain Research, Seattle Children's Research Institute, Washington, 98105, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, 98105, USA
| | - Robert P Doyle
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, 505 Irving Avenue, Syracuse, NY, 13210, USA.
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA.
| | - George G Holz
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, 505 Irving Avenue, Syracuse, NY, 13210, USA.
- Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, 505 Irving Avenue, Syracuse, NY, 13210, USA.
| |
Collapse
|
23
|
Yang R, Wei T, Goldberg H, Wang W, Cullion K, Kohane DS. Getting Drugs Across Biological Barriers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:10.1002/adma.201606596. [PMID: 28752600 PMCID: PMC5683089 DOI: 10.1002/adma.201606596] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 05/30/2017] [Indexed: 05/13/2023]
Abstract
The delivery of drugs to a target site frequently involves crossing biological barriers. The degree and nature of the impediment to flux, as well as the potential approaches to overcoming it, depend on the tissue, the drug, and numerous other factors. Here an overview of approaches that have been taken to crossing biological barriers is presented, with special attention to transdermal drug delivery. Technology and knowledge pertaining to addressing these issues in a variety of organs could have a significant clinical impact.
Collapse
Affiliation(s)
- Rong Yang
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Tuo Wei
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Hannah Goldberg
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Weiping Wang
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Kathleen Cullion
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| |
Collapse
|
24
|
Affiliation(s)
- Young Hee Lee
- Functional Neuroanatomy of Metabolism Regulation Laboratory, Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea.,Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Hee Won Lee
- Functional Neuroanatomy of Metabolism Regulation Laboratory, Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea
| | - Hyung Jin Choi
- Functional Neuroanatomy of Metabolism Regulation Laboratory, Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea.,Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea.,BK21Plus Biomedical Science Project Team, Seoul National University College of Medicine, Seoul, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Wide River Institute of Immunology, Seoul National University, Hongcheon, Korea
| |
Collapse
|
25
|
Gut check on diabesity: leveraging gut mechanisms for the treatment of type 2 diabetes and obesity. Curr Opin Pharmacol 2017; 37:10-15. [PMID: 28802873 DOI: 10.1016/j.coph.2017.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/20/2017] [Indexed: 01/20/2023]
Abstract
Gut hormones have long been understood to regulate food intake and metabolism. Bariatric surgery significantly elevates circulating gut hormone levels and is proven to affect acute remission of type 2 diabetes before any weight loss is observed. Subsequent weight loss is accrued over weeks to months but is sustained into the long term. Hence, there exists great enthusiasm to recapitulate these changes in gut hormones in the form of novel combination drugs for type 2 diabetes and obesity.
Collapse
|
26
|
Rodgers R. Bench to bedside in appetite research: Lost in translation? Neurosci Biobehav Rev 2017; 76:163-173. [DOI: 10.1016/j.neubiorev.2016.08.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/22/2016] [Accepted: 08/24/2016] [Indexed: 12/19/2022]
|
27
|
Abstract
Obesity is a global epidemic that contributes to a number of health complications including cardiovascular disease, type 2 diabetes, cancer and neuropsychiatric disorders. Pharmacotherapeutic strategies to treat obesity are urgently needed. Research over the past two decades has increased substantially our knowledge of central and peripheral mechanisms underlying homeostatic energy balance. Homeostatic mechanisms involve multiple components including neuronal circuits, some originating in hypothalamus and brain stem, as well as peripherally-derived satiety, hunger and adiposity signals that modulate neural activity and regulate eating behavior. Dysregulation of one or more of these homeostatic components results in obesity. Coincident with obesity, reward mechanisms that regulate hedonic aspects of food intake override the homeostatic regulation of eating. In addition to functional interactions between homeostatic and reward systems in the regulation of food intake, homeostatic signals have the ability to alter vulnerability to drug abuse. Regarding the treatment of obesity, pharmacological monotherapies primarily focus on a single protein target. FDA-approved monotherapy options include phentermine (Adipex-P®), orlistat (Xenical®), lorcaserin (Belviq®) and liraglutide (Saxenda®). However, monotherapies have limited efficacy, in part due to the recruitment of alternate and counter-regulatory pathways. Consequently, a multi-target approach may provide greater benefit. Recently, two combination products have been approved by the FDA to treat obesity, including phentermine/topiramate (Qsymia®) and naltrexone/bupropion (Contrave®). The current review provides an overview of homeostatic and reward mechanisms that regulate energy balance, potential therapeutic targets for obesity and current treatment options, including some candidate therapeutics in clinical development. Finally, challenges in anti-obesity drug development are discussed.
Collapse
Affiliation(s)
- Vidya Narayanaswami
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA
| | - Linda P Dwoskin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
28
|
Steinert RE, Feinle-Bisset C, Asarian L, Horowitz M, Beglinger C, Geary N. Ghrelin, CCK, GLP-1, and PYY(3-36): Secretory Controls and Physiological Roles in Eating and Glycemia in Health, Obesity, and After RYGB. Physiol Rev 2017; 97:411-463. [PMID: 28003328 PMCID: PMC6151490 DOI: 10.1152/physrev.00031.2014] [Citation(s) in RCA: 392] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The efficacy of Roux-en-Y gastric-bypass (RYGB) and other bariatric surgeries in the management of obesity and type 2 diabetes mellitus and novel developments in gastrointestinal (GI) endocrinology have renewed interest in the roles of GI hormones in the control of eating, meal-related glycemia, and obesity. Here we review the nutrient-sensing mechanisms that control the secretion of four of these hormones, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide tyrosine tyrosine [PYY(3-36)], and their contributions to the controls of GI motor function, food intake, and meal-related increases in glycemia in healthy-weight and obese persons, as well as in RYGB patients. Their physiological roles as classical endocrine and as locally acting signals are discussed. Gastric emptying, the detection of specific digestive products by small intestinal enteroendocrine cells, and synergistic interactions among different GI loci all contribute to the secretion of ghrelin, CCK, GLP-1, and PYY(3-36). While CCK has been fully established as an endogenous endocrine control of eating in healthy-weight persons, the roles of all four hormones in eating in obese persons and following RYGB are uncertain. Similarly, only GLP-1 clearly contributes to the endocrine control of meal-related glycemia. It is likely that local signaling is involved in these hormones' actions, but methods to determine the physiological status of local signaling effects are lacking. Further research and fresh approaches are required to better understand ghrelin, CCK, GLP-1, and PYY(3-36) physiology; their roles in obesity and bariatric surgery; and their therapeutic potentials.
Collapse
Affiliation(s)
- Robert E Steinert
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christine Feinle-Bisset
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Lori Asarian
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Michael Horowitz
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christoph Beglinger
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Nori Geary
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| |
Collapse
|
29
|
Zaremba SMM, Drummond S, Steinert RE. The need to standardize ad libitum eating protocols in dietary fibre appetite research. Eur J Clin Nutr 2016; 71:570-572. [PMID: 28000690 DOI: 10.1038/ejcn.2016.253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 09/30/2016] [Accepted: 10/26/2016] [Indexed: 11/09/2022]
Affiliation(s)
- S M M Zaremba
- Department of Dietetics, Nutrition and Biological Sciences, Queen Margaret University, Edinburgh, UK
| | - S Drummond
- Department of Dietetics, Nutrition and Biological Sciences, Queen Margaret University, Edinburgh, UK
| | - R E Steinert
- DSM Nutritional Products Ltd. R&D Human Nutrition and Health, Basel, Switzerland
| |
Collapse
|
30
|
Maher S, Mrsny RJ, Brayden DJ. Intestinal permeation enhancers for oral peptide delivery. Adv Drug Deliv Rev 2016; 106:277-319. [PMID: 27320643 DOI: 10.1016/j.addr.2016.06.005] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 12/15/2022]
Abstract
Intestinal permeation enhancers (PEs) are one of the most widely tested strategies to improve oral delivery of therapeutic peptides. This article assesses the intestinal permeation enhancement action of over 250 PEs that have been tested in intestinal delivery models. In depth analysis of pre-clinical data is presented for PEs as components of proprietary delivery systems that have progressed to clinical trials. Given the importance of co-presentation of sufficiently high concentrations of PE and peptide at the small intestinal epithelium, there is an emphasis on studies where PEs have been formulated with poorly permeable molecules in solid dosage forms and lipoidal dispersions.
Collapse
|
31
|
Lakkireddy HR, Urmann M, Besenius M, Werner U, Haack T, Brun P, Alié J, Illel B, Hortala L, Vogel R, Bazile D. Oral delivery of diabetes peptides - Comparing standard formulations incorporating functional excipients and nanotechnologies in the translational context. Adv Drug Deliv Rev 2016; 106:196-222. [PMID: 26964477 DOI: 10.1016/j.addr.2016.02.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/23/2016] [Accepted: 02/28/2016] [Indexed: 12/12/2022]
Abstract
While some orally delivered diabetes peptides are moving to late development with standard formulations incorporating functional excipients, the demonstration of the value of nanotechnology in clinic is still at an early stage. The goal of this review is to compare these two drug delivery approaches from a physico-chemical and a biopharmaceutical standpoint in an attempt to define how nanotechnology-based products can be differentiated from standard oral dosage forms for oral bioavailability of diabetes peptides. Points to consider in a translational approach are outlined to seize the opportunities offered by a better understanding of both the intestinal barrier and of nano-carriers designed for oral delivery.
Collapse
Affiliation(s)
- Harivardhan Reddy Lakkireddy
- Drug Delivery Technologies and Innovation, Pharmaceutical Sciences Operations, Lead Generation and Candidate Realization, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Matthias Urmann
- Diabetes Division, Sanofi Research and Development, Frankfurt, Germany
| | - Melissa Besenius
- Diabetes Division, Sanofi Research and Development, Frankfurt, Germany
| | - Ulrich Werner
- Diabetes Division, Sanofi Research and Development, Frankfurt, Germany
| | - Torsten Haack
- Diabetes Division, Sanofi Research and Development, Frankfurt, Germany
| | - Priscilla Brun
- Disposition Safety and Animal Research, Sanofi Research and Development, Montpellier, France
| | - Jean Alié
- Analytical Sciences, Lead Generation and Candidate Realization, Sanofi Research and Development, Montpellier, France
| | - Brigitte Illel
- Pharmaceutical Sciences Operations, Lead Generation and Candidate Realization, Sanofi Research and Development, Montpellier, France
| | - Laurent Hortala
- Pharmaceutical Sciences Operations, Lead Generation and Candidate Realization, Sanofi Research and Development, Montpellier, France
| | - Rachel Vogel
- Pharmaceutical Sciences Operations, Lead Generation and Candidate Realization, Sanofi Research and Development, Montpellier, France
| | - Didier Bazile
- Drug Delivery Technologies and Innovation, Pharmaceutical Sciences Operations, Lead Generation and Candidate Realization, Sanofi Research and Development, Vitry-sur-Seine, France.
| |
Collapse
|
32
|
Schober G, Lange K, Steinert RE, Hutchison AT, Luscombe-Marsh ND, Landrock MF, Horowitz M, Seimon RV, Feinle-Bisset C. Contributions of upper gut hormones and motility to the energy intake-suppressant effects of intraduodenal nutrients in healthy, lean men - a pooled-data analysis. Physiol Rep 2016; 4:e12943. [PMID: 27613824 PMCID: PMC5027351 DOI: 10.14814/phy2.12943] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/08/2016] [Indexed: 02/07/2023] Open
Abstract
We have previously identified pyloric pressures and plasma cholecystokinin (CCK) concentrations as independent determinants of energy intake following administration of intraduodenal lipid and intravenous CCK. We evaluated in healthy men whether these parameters also determine energy intake in response to intraduodenal protein, and whether, across the nutrients, any predominant gastrointestinal (GI) factors exist, or many factors make small contributions. Data from nine published studies, in which antropyloroduodenal pressures, GI hormones, and GI /appetite perceptions were measured during intraduodenal lipid or protein infusions, were pooled. In all studies energy intake was quantified immediately after the infusions. Specific variables for inclusion in a mixed-effects multivariable model for determination of independent predictors of energy intake were chosen following assessment for collinearity, and within-subject correlations between energy intake and these variables were determined using bivariate analyses adjusted for repeated measures. In models based on all studies, or lipid studies, there were significant effects for amplitude of antral pressure waves, premeal glucagon-like peptide-1 (GLP-1) and time-to-peak GLP-1 concentrations, GLP-1 AUC and bloating scores (P < 0.05), and trends for basal pyloric pressure (BPP), amplitude of duodenal pressure waves, peak CCK concentrations, and hunger and nausea scores (0.05 < P ≤ 0.094), to be independent determinants of subsequent energy intake. In the model including the protein studies, only BPP was identified as an independent determinant of energy intake (P < 0.05). No single parameter was identified across all models, and effects of the variables identified were relatively small. Taken together, while GI mechanisms contribute to the regulation of acute energy intake by lipid and protein, their contribution to the latter is much less. Moreover, the effects are likely to reflect small, cumulative contributions from a range of interrelated factors.
Collapse
Affiliation(s)
- Gudrun Schober
- University of Adelaide Discipline of Medicine, Adelaide, Australia
| | - Kylie Lange
- University of Adelaide Discipline of Medicine, Adelaide, Australia NHMRC Centre of Excellence in Translating Nutritional Science to Good Health University of Adelaide, Adelaide, Australia
| | - Robert E Steinert
- University of Adelaide Discipline of Medicine, Adelaide, Australia NHMRC Centre of Excellence in Translating Nutritional Science to Good Health University of Adelaide, Adelaide, Australia
| | - Amy T Hutchison
- University of Adelaide Discipline of Medicine, Adelaide, Australia NHMRC Centre of Excellence in Translating Nutritional Science to Good Health University of Adelaide, Adelaide, Australia South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Natalie D Luscombe-Marsh
- NHMRC Centre of Excellence in Translating Nutritional Science to Good Health University of Adelaide, Adelaide, Australia CSIRO Animal, Food and Health Sciences, Adelaide, Australia
| | - Maria F Landrock
- University of Adelaide Discipline of Medicine, Adelaide, Australia
| | - Michael Horowitz
- University of Adelaide Discipline of Medicine, Adelaide, Australia NHMRC Centre of Excellence in Translating Nutritional Science to Good Health University of Adelaide, Adelaide, Australia
| | - Radhika V Seimon
- Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, University of Sydney, Sydney, Australia
| | - Christine Feinle-Bisset
- University of Adelaide Discipline of Medicine, Adelaide, Australia NHMRC Centre of Excellence in Translating Nutritional Science to Good Health University of Adelaide, Adelaide, Australia
| |
Collapse
|
33
|
Oral delivery of macromolecular drugs: Where we are after almost 100years of attempts. Adv Drug Deliv Rev 2016; 101:108-121. [PMID: 26826437 DOI: 10.1016/j.addr.2016.01.010] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 01/11/2016] [Accepted: 01/18/2016] [Indexed: 02/08/2023]
Abstract
Since the first attempt to administer insulin orally in humans more than 90years ago, the oral delivery of macromolecular drugs (>1000g/mol) has been rather disappointing. Although several clinical pilot studies have demonstrated that the oral absorption of macromolecules is possible, the bioavailability remains generally low and variable. This article reviews the formulations and biopharmaceutical aspects of orally administered biomacromolecules on the market and in clinical development for local and systemic delivery. The most successful approaches for systemic delivery often involve a combination of enteric coating, protease inhibitors and permeation enhancers in relatively high amounts. However, some of these excipients have induced local or systemic adverse reactions in preclinical and clinical studies, and long-term studies are often missing. Therefore, strategies aimed at increasing the oral absorption of macromolecular drugs should carefully take into account the benefit-risk ratio. In the absence of specific uptake pathways, small and potent peptides that are resistant to degradation and that present a large therapeutic window certainly represent the best candidates for systemic absorption. While we acknowledge the need for systemically delivering biomacromolecules, it is our opinion that the oral delivery to local gastrointestinal targets is currently more promising because of their accessibility and the lacking requirement for intestinal permeability enhancement.
Collapse
|
34
|
Teuffel P, Goebel-Stengel M, Hofmann T, Prinz P, Scharner S, Körner JL, Grötzinger C, Rose M, Klapp BF, Stengel A. A RAPID Method for Blood Processing to Increase the Yield of Plasma Peptide Levels in Human Blood. J Vis Exp 2016. [PMID: 27166969 DOI: 10.3791/53959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Research in the field of food intake regulation is gaining importance. This often includes the measurement of peptides regulating food intake. For the correct determination of a peptide's concentration, it should be stable during blood processing. However, this is not the case for several peptides which are quickly degraded by endogenous peptidases. Recently, we developed a blood processing method employing Reduced temperatures, Acidification, Protease inhibition, Isotopic exogenous controls and Dilution (RAPID) for the use in rats. Here, we have established this technique for the use in humans and investigated recovery, molecular form and circulating concentration of food intake regulatory hormones. The RAPID method significantly improved the recovery for (125)I-labeled somatostatin-28 (+39%), glucagon-like peptide-1 (+35%), acyl ghrelin and glucagon (+32%), insulin and kisspeptin (+29%), nesfatin-1 (+28%), leptin (+21%) and peptide YY3-36 (+19%) compared to standard processing (EDTA blood on ice, p <0.001). High performance liquid chromatography showed the elution of endogenous acyl ghrelin at the expected position after RAPID processing, while after standard processing 62% of acyl ghrelin were degraded resulting in an earlier peak likely representing desacyl ghrelin. After RAPID processing the acyl/desacyl ghrelin ratio in blood of normal weight subjects was 1:3 compared to 1:23 following standard processing (p = 0.03). Also endogenous kisspeptin levels were higher after RAPID compared to standard processing (+99%, p = 0.02). The RAPID blood processing method can be used in humans, yields higher peptide levels and allows for assessment of the correct molecular form.
Collapse
Affiliation(s)
- Pauline Teuffel
- Charité Center for Internal Medicine and Dermatology, Division General Internal and Psychosomatic Medicine, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin
| | - Miriam Goebel-Stengel
- Charité Center for Internal Medicine and Dermatology, Division General Internal and Psychosomatic Medicine, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin; Department of Internal Medicine, Institute of Neurogastroenterology and Motility, Martin-Luther Hospital, Academic Teaching Institution of Charité Universitätsmedizin Berlin
| | - Tobias Hofmann
- Charité Center for Internal Medicine and Dermatology, Division General Internal and Psychosomatic Medicine, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin
| | - Philip Prinz
- Charité Center for Internal Medicine and Dermatology, Division General Internal and Psychosomatic Medicine, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin
| | - Sophie Scharner
- Charité Center for Internal Medicine and Dermatology, Division General Internal and Psychosomatic Medicine, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin
| | - Jan L Körner
- Department of Hepatology and Gastroenterology, Molecular Cancer Research Center (MKFZ), Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin
| | - Carsten Grötzinger
- Department of Hepatology and Gastroenterology, Molecular Cancer Research Center (MKFZ), Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin
| | - Matthias Rose
- Charité Center for Internal Medicine and Dermatology, Division General Internal and Psychosomatic Medicine, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin
| | - Burghard F Klapp
- Charité Center for Internal Medicine and Dermatology, Division General Internal and Psychosomatic Medicine, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Division General Internal and Psychosomatic Medicine, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin;
| |
Collapse
|
35
|
Grunddal KV, Ratner CF, Svendsen B, Sommer F, Engelstoft MS, Madsen AN, Pedersen J, Nøhr MK, Egerod KL, Nawrocki AR, Kowalski T, Howard AD, Poulsen SS, Offermanns S, Bäckhed F, Holst JJ, Holst B, Schwartz TW. Neurotensin Is Coexpressed, Coreleased, and Acts Together With GLP-1 and PYY in Enteroendocrine Control of Metabolism. Endocrinology 2016; 157:176-94. [PMID: 26469136 DOI: 10.1210/en.2015-1600] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The 2 gut hormones glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) are well known to be coexpressed, costored, and released together to coact in the control of key metabolic target organs. However, recently, it became clear that several other gut hormones can be coexpressed in the intestinal-specific lineage of enteroendocrine cells. Here, we focus on the anatomical and functional consequences of the coexpression of neurotensin with GLP-1 and PYY in the distal small intestine. Fluorescence-activated cell sorting analysis, laser capture, and triple staining demonstrated that GLP-1 cells in the crypts become increasingly multihormonal, ie, coexpressing PYY and neurotensin as they move up the villus. Proglucagon promoter and pertussis toxin receptor-driven cell ablation and reappearance studies indicated that although all the cells die, the GLP-1 cells reappear more quickly than PYY- and neurotensin-positive cells. High-resolution confocal fluorescence microscopy demonstrated that neurotensin is stored in secretory granules distinct from GLP-1 and PYY storing granules. Nevertheless, the 3 peptides were cosecreted from both perfused small intestines and colonic crypt cultures in response to a series of metabolite, neuropeptide, and hormonal stimuli. Importantly, neurotensin acts synergistically, ie, more than additively together with GLP-1 and PYY to decrease palatable food intake and inhibit gastric emptying, but affects glucose homeostasis in a more complex manner. Thus, neurotensin is a major gut hormone deeply integrated with GLP-1 and PYY, which should be taken into account when exploiting the enteroendocrine regulation of metabolism pharmacologically.
Collapse
Affiliation(s)
- Kaare V Grunddal
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Cecilia F Ratner
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Berit Svendsen
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Felix Sommer
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Maja S Engelstoft
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Andreas N Madsen
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Jens Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Mark K Nøhr
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Kristoffer L Egerod
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Andrea R Nawrocki
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Timothy Kowalski
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Andrew D Howard
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Steen Seier Poulsen
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Stefan Offermanns
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Fredrik Bäckhed
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Birgitte Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thue W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research (K.V.G., C.F.R., B.S., M.S.E., A.N.M., J.P., M.K.N., K.L.E., F.B., J.J.H., B.H., T.W.S.), Section for Metabolic Receptology and Enteroendocrinology; Laboratory for Molecular Pharmacology (K.V.G., C.F.R., M.S.E., A.N.M., M.K.N., K.L.E., B.H., T.W.S.), Department of Neuroscience and Pharmacology; and Department of Biomedical Sciences (B.S., J.P., S.S.P., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Department of Molecular and Clinical Medicine (F.S., F.B.), Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Danish Diabetes Academy (M.S.E.), 5000 Odense, Denmark; Merck Research Laboratories (A.R.N., T.K., A.D.H.), Kenilworth, NJ 07033; and Department of Pharmacology (S.O.), Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
36
|
Intestinal GLP-1 and satiation: from man to rodents and back. Int J Obes (Lond) 2015; 40:198-205. [PMID: 26315842 DOI: 10.1038/ijo.2015.172] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 08/27/2015] [Accepted: 08/12/2015] [Indexed: 02/06/2023]
Abstract
In response to luminal food stimuli during meals, enteroendocrine cells release gastrointestinal (GI) peptides that have long been known to control secretory and motor functions of the gut, pancreas and liver. Glucagon-like peptide-1 (GLP-1) has emerged as one of the most important GI peptides because of a combination of functions not previously ascribed to any other molecule. GLP-1 potentiates glucose-induced insulin secretion, suppresses glucagon release, slows gastric emptying and may serve as a satiation signal, although the physiological status of the latter function has not been fully established yet. Here we review the available evidence for intestinal GLP-1 to fulfill a number of established empirical criteria for assessing whether a hormone inhibits eating by eliciting physiological satiation in man and rodents.
Collapse
|
37
|
He W, Parowatkin M, Mailänder V, Flechtner-Mors M, Graf R, Best A, Koynov K, Mohr K, Ziener U, Landfester K, Crespy D. Nanocarrier for Oral Peptide Delivery Produced by Polyelectrolyte Complexation in Nanoconfinement. Biomacromolecules 2015; 16:2282-7. [DOI: 10.1021/acs.biomac.5b00500] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Wei He
- Max Planck Institute
for Polymer Research, Ackermannweg
10, 55128, Mainz, Germany
| | - Maria Parowatkin
- Max Planck Institute
for Polymer Research, Ackermannweg
10, 55128, Mainz, Germany
| | - Volker Mailänder
- Max Planck Institute
for Polymer Research, Ackermannweg
10, 55128, Mainz, Germany
| | - Marion Flechtner-Mors
- University of Ulm, Institute of Micro- and Nanomaterials, Albert-Einstein-Allee 47, D-89081, Ulm, Germany
| | - Robert Graf
- Max Planck Institute
for Polymer Research, Ackermannweg
10, 55128, Mainz, Germany
| | - Andreas Best
- Max Planck Institute
for Polymer Research, Ackermannweg
10, 55128, Mainz, Germany
| | - Kaloian Koynov
- Max Planck Institute
for Polymer Research, Ackermannweg
10, 55128, Mainz, Germany
| | - Kristin Mohr
- Max Planck Institute
for Polymer Research, Ackermannweg
10, 55128, Mainz, Germany
| | - Ulrich Ziener
- University of Ulm, Institute of Organic Chemistry III, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Katharina Landfester
- Max Planck Institute
for Polymer Research, Ackermannweg
10, 55128, Mainz, Germany
| | - Daniel Crespy
- Max Planck Institute
for Polymer Research, Ackermannweg
10, 55128, Mainz, Germany
| |
Collapse
|
38
|
Karsdal MA, Riis BJ, Mehta N, Stern W, Arbit E, Christiansen C, Henriksen K. Lessons learned from the clinical development of oral peptides. Br J Clin Pharmacol 2015; 79:720-32. [PMID: 25408230 PMCID: PMC4415709 DOI: 10.1111/bcp.12557] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/14/2014] [Indexed: 01/11/2023] Open
Abstract
The oral delivery of peptides and proteins has been hampered by an array of obstacles. However, several promising novel oral delivery systems have been developed. This paper reviews the most advanced oral formulation technologies, and highlights key lessons and implications from studies undertaken to date with these oral formulations. Special interest is given to oral salmon calcitonin (CT), glucagon-like peptide-1 (GLP-1), insulin, PYY-(3-36), recombinant human parathyroid hormone (rhPTH(1-31)-NH2) and PTH(1-34), by different technologies. The issues addressed include (i) interaction with water, (ii) interaction with food, (iii) diurnal variation, (iv) inter- and intra-subject variability, (v) correlation between efficacy and exposure and (vi) key deliverables of different technologies. These key lessons may aid research in the development of other oral formulations.
Collapse
|
39
|
Scott RV, Tan TM, Bloom SR. Can Bayliss and Starling gut hormones cure a worldwide pandemic? J Physiol 2014; 592:5153-67. [PMID: 25217372 PMCID: PMC4262331 DOI: 10.1113/jphysiol.2014.272955] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/04/2014] [Indexed: 12/17/2022] Open
Abstract
Bayliss and Starling first coined the term 'hormone' with reference to secretin, a substance they found that was produced by the gut, but released into the blood stream to act at a distance. The intestine is now known as the largest endocrine organ in the body, and it produces numerous hormones with a wide range of functions. These include controlling appetite and energy homeostasis. Obesity is one of the greatest health threats facing the world today. At present, the only successful treatment is surgery. Bariatric procedures such as the Roux-en-Y bypass work by elevating gut hormones that induce satiety. Significant research has gone into producing versions of these hormones that can be delivered therapeutically to treat obesity. This review looks at the role of gut hormones in obesity, and the development of gut hormone-derived obesity treatments.
Collapse
Affiliation(s)
- R V Scott
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - T M Tan
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - S R Bloom
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
40
|
Finelli C, Padula MC, Martelli G, Tarantino G. Could the improvement of obesity-related co-morbidities depend on modified gut hormones secretion? World J Gastroenterol 2014; 20:16649-16664. [PMID: 25469034 PMCID: PMC4248209 DOI: 10.3748/wjg.v20.i44.16649] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 05/27/2014] [Accepted: 06/25/2014] [Indexed: 02/06/2023] Open
Abstract
Obesity and its associated diseases are a worldwide epidemic disease. Usual weight loss cures - as diets, physical activity, behavior therapy and pharmacotherapy - have been continuously implemented but still have relatively poor long-term success and mainly scarce adherence. Bariatric surgery is to date the most effective long term treatment for morbid obesity and it has been proven to reduce obesity-related co-morbidities, among them nonalcoholic fatty liver disease, and mortality. This article summarizes such variations in gut hormones following the current metabolic surgery procedures. The profile of gut hormonal changes after bariatric surgery represents a strategy for the individuation of the most performing surgical procedures to achieve clinical results. About this topic, experts suggest that the individuation of the crosslink among the gut hormones, microbiome, the obesity and the bariatric surgery could lead to new and more specific therapeutic interventions for severe obesity and its co-morbidities, also non surgical.
Collapse
|
41
|
Valsamakis G, Lois K, Kumar S, Mastorakos G. New molecular targets in the pathophysiology of obesity and available treatment options under investigation. Clin Obes 2014; 4:209-19. [PMID: 25826792 DOI: 10.1111/cob.12064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 05/12/2014] [Accepted: 05/14/2014] [Indexed: 12/22/2022]
Abstract
The pharmacotherapy of obesity has historically recorded an overall poor safety and efficacy profile largely because of the complex mechanisms involved in the pathophysiology of obesity. It is hoped that a better understanding of the regulation of body weight will lead us to the development of effective and safer drugs. Recent advances in our understanding of the regulation of energy homeostasis has allowed the design of novel anti-obesity drugs targeting specific molecules crucial for the modulation of energy balance, including drugs that induce satiety, modulate nutrient absorption or influence metabolism or lipogenesis. Almost a decade after the Food and Drug Administration approved the first weight loss medication, it recently approved two novel anti-obesity drugs Belviq (lorcaserin) and Qsymia (topiramate and phentermine), thus signalling the beginning of a new era in the pharmacotherapy of obesity. It is believed that the next generation of weight-loss drugs will be based on combination treatments with gut hormones in a manner that mimics the changes underlying surgically induced weight loss thus introducing the so called 'bariatric pharmacotherapy'. An in-depth understanding of the interrelated physiological and behavioural effects of these new molecules together with the development of new treatment paradigms is needed so that future disappointments in the field of obesity pharmacotherapy may be avoided.
Collapse
Affiliation(s)
- G Valsamakis
- Endocrine Unit, 2nd Department of Obs and Gynae, Areteeion University Hospital, Athens Medical School National and Kapodistrian University of Athens, Athens, Greece; WISDEM Centre for Diabetes, Endocrinology and Metabolism, Warwick Medical School, University of Warwick, Coventry, UK
| | | | | | | |
Collapse
|
42
|
|
43
|
Abstract
As obesity continues to be a global epidemic, research into the mechanisms of hunger and satiety and how those signals act to regulate energy homeostasis persists. Peptide YY (PYY) is an acute satiety signal released upon nutrient ingestion and has been shown to decrease food intake when administered exogenously. More recently, investigators have studied how different factors influence PYY release and circulating levels in humans. Some of these factors include exercise, macronutrient composition of the diet, body-weight status, adiposity levels, sex, race and ageing. The present article provides a succinct and comprehensive review of the recent literature published on the different factors that influence PYY release and circulating levels in humans. Where human data are insufficient, evidence in animal or cell models is summarised. Additionally, the present review explores the recent findings on PYY responses to different dietary fatty acids and how this new line of research will make an impact on future studies on PYY. Human demographics, such as sex and age, do not appear to influence PYY levels. Conversely, adiposity or BMI, race and acute exercise all influence circulating PYY levels. Both dietary fat and protein strongly stimulate PYY release. Furthermore, MUFA appear to result in a smaller PYY response compared with SFA and PUFA. PYY levels appear to be affected by acute exercise, macronutrient composition, adiposity, race and the composition of fatty acids from dietary fat.
Collapse
|
44
|
Schmidt JB, Gregersen NT, Pedersen SD, Arentoft JL, Ritz C, Schwartz TW, Holst JJ, Astrup A, Sjödin A. Effects of PYY3-36 and GLP-1 on energy intake, energy expenditure, and appetite in overweight men. Am J Physiol Endocrinol Metab 2014; 306:E1248-56. [PMID: 24735885 DOI: 10.1152/ajpendo.00569.2013] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Our aim was to examine the effects of GLP-1 and PYY3-36, separately and in combination, on energy intake, energy expenditure, appetite sensations, glucose and fat metabolism, ghrelin, and vital signs in healthy overweight men. Twenty-five healthy male subjects participated in this randomized, double-blinded, placebo-controlled, four-arm crossover study (BMI 29 ± 3 kg/m(2), age 33 ± 9 yr). On separate days they received a 150-min intravenous infusion of 1) 0.8 pmol·kg(-1)·min(-1) PYY3-36, 2) 1.0 pmol·kg(-1)·min(-1) GLP-1, 3) GLP-1 + PYY3-36, or 4) placebo. Ad libitum energy intake was assessed during the final 30 min. Measurements of appetite sensations, energy expenditure and fat oxidation, vital signs, and blood variables were collected throughout the infusion period. No effect on energy intake was found after monoinfusions of PYY3-36 (-4.2 ± 4.8%, P = 0.8) or GLP-1 (-3.0 ± 4.5%, P = 0.9). However, the coinfusion reduced energy intake compared with placebo (-30.4 ± 6.5%, P < 0.0001) and more than the sum of the monoinfusions (P < 0.001), demonstrating a synergistic effect. Coinfusion slightly increased sensation of nausea (P < 0.05), but this effect could not explain the effect on energy intake. A decrease in plasma ghrelin was found after all treatments compared with placebo (all P < 0.05); however, infusions of GLP-1 + PYY3-36 resulted in an additional decrease compared with the monoinfusions (both P < 0.01). We conclude that coinfusion of GLP-1 and PYY3-36 exerted a synergistic effect on energy intake. The satiating effect of the meal was enhanced by GLP-1 and PYY3-36 in combination compared with placebo. Coinfusion was accompanied by slightly increased nausea and a decrease in plasma ghrelin, but neither of these factors could explain the reduction in energy intake.
Collapse
Affiliation(s)
- Julie Berg Schmidt
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark;
| | - Nikolaj Ture Gregersen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Sue D Pedersen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Johanne L Arentoft
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Christian Ritz
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Thue W Schwartz
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Novo Nordisk Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Anders Sjödin
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
45
|
Abstract
Delivery of peptides by the oral route greatly appeals due to commercial, patient convenience and scientific arguments. While there are over 60 injectable peptides marketed worldwide, and many more in development, most delivery strategies do not yet adequately overcome the barriers to oral delivery. Peptides are sensitive to chemical and enzymatic degradation in the intestine, and are poorly permeable across the intestinal epithelium due to sub-optimal physicochemical properties. A successful oral peptide delivery technology should protect potent peptides from presystemic degradation and improve epithelial permeation to achieve a target oral bioavailability with acceptable intra-subject variability. This review provides a comprehensive up-to-date overview of the current status of oral peptide delivery with an emphasis on patented formulations that are yielding promising clinical data.
Collapse
|
46
|
Punjabi M, Arnold M, Rüttimann E, Graber M, Geary N, Pacheco-López G, Langhans W. Circulating glucagon-like peptide-1 (GLP-1) inhibits eating in male rats by acting in the hindbrain and without inducing avoidance. Endocrinology 2014; 155:1690-9. [PMID: 24601880 DOI: 10.1210/en.2013-1447] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
To address the neural mediation of the eating-inhibitory effect of circulating glucagon-like peptide-1 (GLP-1), we investigated the effects of 1) intra-fourth ventricular infusion of the GLP-1 receptor antagonist exendin-9 or 2) area postrema lesion on the eating-inhibitory effect of intrameal hepatic portal vein (HPV) GLP-1 infusion in adult male rats. To evaluate the physiological relevance of the observed effect we examined 3) the influence of GLP-1 on flavor acceptance in a 2-bottle conditioned flavor avoidance test, and 4) measured active GLP-1 in the HPV and vena cava (VC) in relation to a meal and in the VC after HPV GLP-1 infusion. Intrameal HPV GLP-1 infusion (1 nmol/kg body weight-5 min) specifically reduced ongoing meal size by almost 40% (P < .05). Intra-fourth ventricular exendin-9 (10 μg/rat) itself did not affect eating, but attenuated (P < .05) the satiating effect of HPV GLP-1. Area postrema lesion also blocked (P < .05) the eating-inhibitory effect of HPV GLP-1. Pairing consumption of flavored saccharin solutions with HPV GLP-1 infusion did not alter flavor acceptance, indicating that HPV GLP-1 can inhibit eating without inducing malaise. A regular chow meal transiently increased (P < .05) HPV, but not VC, plasma active GLP-1 levels, whereas HPV GLP-1 infusion caused a transient supraphysiological increase (P < .01) in VC GLP-1 concentration 3 minutes after infusion onset. The results implicate hindbrain GLP-1 receptors and the area postrema in the eating-inhibitory effect of circulating GLP-1, but question the physiological relevance of the eating-inhibitory effect of iv infused GLP-1 under our conditions.
Collapse
Affiliation(s)
- Mukesh Punjabi
- Physiology and Behavior Laboratory, Institute of Food, Nutrition, and Health, Swiss Federal Institute of Technology Zurich (ETH Zurich), 8603 Schwerzenbach, Switzerland
| | | | | | | | | | | | | |
Collapse
|
47
|
Troke RC, Tan TM, Bloom SR. The future role of gut hormones in the treatment of obesity. Ther Adv Chronic Dis 2014; 5:4-14. [PMID: 24381724 PMCID: PMC3871274 DOI: 10.1177/2040622313506730] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The obesity pandemic presents a significant burden, both in terms of healthcare and economic outcomes, and current medical therapies are inadequate to deal with this challenge. Bariatric surgery is currently the only therapy available for obesity which results in long-term, sustained weight loss. The favourable effects of this surgery are thought, at least in part, to be mediated via the changes of gut hormones such as GLP-1, PYY, PP and oxyntomodulin seen following the procedure. These hormones have subsequently become attractive novel targets for the development of obesity therapies. Here, we review the development of these gut peptides as current and emerging therapies in the treatment of obesity.
Collapse
Affiliation(s)
- Rachel C Troke
- Department of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Tricia M Tan
- Department of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Steve R Bloom
- Department of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, 6th Floor, Commonwealth Building, London W12 0HS, UK
| |
Collapse
|
48
|
Siekmeier R, Hofmann T, Scheuch G, Pokorski M. Aerosolized GLP-1 for treatment of diabetes mellitus and irritable bowel syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 849:23-38. [PMID: 25427821 DOI: 10.1007/5584_2014_94] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Diabetes is a global burden and the prevalence of the disease, in particular diabetes mellitus type 2 is rapidly increasing worldwide. After introduction of insulin into clinical therapy about 90 years ago a major number of pharmaceuticals has been developed for treatment of diabetes mellitus type 2. One of these, the incretin glucagon-like peptide 1 (GLP-1), like insulin, needs subcutaneous administration causing inconvenience to patients. However, administration of GLP-1 plays also a role for treatment of irritable bowel syndrome (IBS). To improve patient convenience inhaled insulin (Exubera(®)) was developed and approved but failed market acceptance some years ago. Recently, another inhalative insulin (Afrezza(®)) received market approval and GLP-1 may serve as another candidate drug for inhalative administration. This review analyzes the current literature investigating alternative administration of GLP-1 and GLP-1 analogs focusing on inhalation. Several formulations for inhalative administration of GLP-1 and analogs were investigated in animal studies, whereas there are only few clinical data. However, feasibility of GLP-1 inhalation has been shown and should be further investigated as such type of drug administration may serve for improvement of therapy in patients with diabetes mellitus or irritable bowel syndrome.
Collapse
Affiliation(s)
- Rüdiger Siekmeier
- Drug Regulatory Affairs, Pharmaceutical Institute, Bonn University, An der Immenburg 4, 53121, Bonn, Germany,
| | | | | | | |
Collapse
|
49
|
Marathe CS, Rayner CK, Jones KL, Horowitz M. Glucagon-like peptides 1 and 2 in health and disease: a review. Peptides 2013; 44:75-86. [PMID: 23523778 DOI: 10.1016/j.peptides.2013.01.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 01/30/2013] [Accepted: 01/30/2013] [Indexed: 02/07/2023]
Abstract
The gut derived peptides, glucagon-like peptides 1 and 2 (GLP-1 and GLP-2), are secreted following nutrient ingestion. GLP-1 and another gut peptide, glucose-dependent insulinotropic polypeptide (GIP) are collectively referred to as 'incretin' hormones, and play an important role in glucose homeostasis. Incretin secretion shares a complex interdependent relationship with both postprandial glycemia and the rate of gastric emptying. GLP-1 based therapies are now well established in the management of type 2 diabetes, while recent literature has suggested potential applications to treat obesity and protect against cardiovascular and neurological disease. The mechanism of action of GLP-2 is not well understood, but it shows promise as an intestinotropic agent.
Collapse
Affiliation(s)
- Chinmay S Marathe
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia.
| | | | | | | |
Collapse
|
50
|
Kim GW, Lin JE, Blomain ES, Waldman SA. New advances in models and strategies for developing anti-obesity drugs. Expert Opin Drug Discov 2013; 8:655-71. [PMID: 23621300 DOI: 10.1517/17460441.2013.792804] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Obesity is a worldwide pandemic. Obesity-related health and economic costs are staggering. Existing strategies to combat obesity through lifestyle improvements and medical intervention have had limited success. Pharmacotherapy, in combination with lifestyle modification, may play a vital role in reversing the disease burden. However, past and current weight-loss medications have had serious safety risks, notably cardiovascular and psychiatric events. AREAS COVERED The authors review the strategies for designing new anti-obesity drugs by describing those currently in development. They describe their target, mechanism of action and developmental or regulatory status. Furthermore, they discuss the problem of weight regain following weight loss, and its relevance to the long-term success of anti-obesity pharmacotherapy. EXPERT OPINION For weight management drugs to achieve the safety and efficacy required to be impactful, current studies are uncovering and characterizing new targets, including new signaling circuits and hormones regulating appetite and metabolism, and re-evaluating the role of pharmacotherapy in weight management. To avoid the safety failures of many past weight-loss drugs, the models and strategies covered in this article incorporate recent advances in knowledge and technology. We discuss the emergence of cGMP signaling as a potentially transformative target in weight management. Modulating cGMP signaling may represent an ideal goal for an anti-obesity pharmacotherapy, reflecting some of the major themes described in the present review: targeting pathways that are newly realized as relevant for weight management; promoting safety by re-purposing drugs that are safe, proven, and approved for clinical use; and having a synergistic effect on multiple, reinforcing pathways.
Collapse
Affiliation(s)
- Gilbert W Kim
- Thomas Jefferson University, Department of Pharmacology and Experimental Therapeutics, Philadelphia, PA 19107, USA
| | | | | | | |
Collapse
|