1
|
Xu Y, Kuipers OP. Design and Biosynthesis of Ornithine 8-Containing Semaglutide Variants with a Click Chemistry-Modifiable Position 26. ACS Synth Biol 2025; 14:1790-1801. [PMID: 40305415 PMCID: PMC12090216 DOI: 10.1021/acssynbio.5c00132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 05/02/2025]
Abstract
Semaglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, constitutes an effective and widely used treatment for type 2 diabetes and obesity. However, challenges such as insufficient oral bioavailability, gastrointestinal side effects, and high costs persist. Overcoming these limitations is essential for improving patient compliance and semaglutide's safety profile. While advanced technologies such as oral delivery systems offer partial solutions, optimizing the peptide structure is crucial for addressing these issues. Establishing a rapid method to generate a large library of semaglutide mutants will enable high-throughput activity screening. In this study, we introduce a novel "Fits-In-All" approach that combines ribosomally synthesized and post-translationally modified peptide (RiPP) technology with amber stop codon incorporation to generate semaglutide variants. To counter dipeptidyl peptidase-4-mediated cleavage, our method strategically incorporates noncanonical amino acid ornithine at position 8 utilizing microbial modification enzyme OspR in vivo. Furthermore, functional groups are introduced by an orthogonal tRNA/aminoacyl-tRNA synthetase pair recognizing the amber stop codon at position 26, which enabled the click chemistry-based linkage of diverse groups. This approach allows for the generation of a broad array of semaglutide analogues that can be screened for optimal properties. In conclusion, this innovative approach opens new avenues for the design and synthesis of optimized peptide-based GLP-1 receptor agonists.
Collapse
Affiliation(s)
- Yanli Xu
- Department of Molecular Genetics,
Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen 9747 AG, The Netherlands
| | - Oscar P. Kuipers
- Department of Molecular Genetics,
Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen 9747 AG, The Netherlands
| |
Collapse
|
2
|
Persson C, Eaton A, Mayrovitz HN. A Closer Look at the Dermatological Profile of GLP-1 Agonists. Diseases 2025; 13:127. [PMID: 40422559 DOI: 10.3390/diseases13050127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND/OBJECTIVES Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are widely used in treating type 2 diabetes and obesity, offering established metabolic and cardiovascular benefits. Emerging evidence suggests these agents also exert direct dermatologic effects. This systematic review categorizes these effects and explores their role in inflammatory skin diseases. METHODS A comprehensive literature search was performed across EMBASE, PubMed, Web of Science, and Google Scholar for studies published from 2014 to 2025. Inclusion criteria were English-language, peer-reviewed original research involving human subjects that linked GLP-1RAs to dermatologic effects. Animal and in vitro studies were excluded. PRISMA guidelines were followed. RESULTS Fifty-one studies met inclusion criteria. Thirty-four reported adverse effects, including hypersensitivity, injection-site reactions, pruritus, urticaria, angioedema, and immune-mediated conditions like bullous pemphigoid. Seventeen studies described beneficial outcomes, such as improvements in psoriasis, reduced hidradenitis suppurativa flares, enhanced wound healing, anti-aging potential, and decreased inflammation. GLP-1RAs showed cytokine modulation in psoriasis, though their role in hidradenitis suppurativa remains uncertain. Cosmetic concerns, such as "Ozempic Face" due to rapid weight loss, were also noted. CONCLUSIONS GLP-1RAs have a broad spectrum of dermatologic effects, from immunomodulatory benefits to adverse cutaneous reactions. Their impact on inflammatory skin disorders suggests a novel therapeutic avenue. However, adverse reactions and aesthetic changes warrant vigilance. Future research should focus on mechanistic studies, long-term safety, and identifying biomarkers to predict dermatologic responses, ultimately guiding personalized treatment approaches.
Collapse
Affiliation(s)
- Calista Persson
- Dr. Kiran Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, FL 33328, USA
| | - Allison Eaton
- Dr. Kiran Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, FL 33328, USA
| | - Harvey N Mayrovitz
- Dr. Kiran Patel College of Allopathic Medicine, Nova Southeastern University, Davie, FL 33314, USA
| |
Collapse
|
3
|
Singh SS, Calvo R, Kumari A, Sable RV, Fang Y, Tao D, Hu X, Castle SG, Nahar S, Li D, Major E, Sanchez TW, Kato R, Xu X, Zhou J, Liu L, LeClair CA, Simeonov A, Baljinnyam B, Henderson MJ, Marugan J, Rudloff U. Fatty Acid Derivatization and Cyclization of the Immunomodulatory Peptide RP-182 Targeting CD206high Macrophages Improve Antitumor Activity. Mol Cancer Ther 2024; 23:1827-1841. [PMID: 39212669 PMCID: PMC11612619 DOI: 10.1158/1535-7163.mct-23-0790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 06/04/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
As tumor-associated macrophages (TAM) exercise a plethora of protumor and immune evasive functions, novel strategies targeting TAMs to inhibit tumor progression have emerged within the current arena of cancer immunotherapy. Activation of the mannose receptor 1 (CD206) is a recent approach that recognizes immunosuppressive CD206high M2-like TAMs as a drug target. Ligation of CD206 both induces reprogramming of CD206high TAMs toward a proinflammatory phenotype and selectively triggers apoptosis in these cells. CD206-activating therapeutics are currently limited to the linear, 10mer peptide RP-182, 1, which is not a drug candidate. In this study, we sought to identify a better suitable candidate for future clinical development by synthesizing and evaluating a series of RP-182 analogs. Surprisingly, fatty acid derivative 1a [RP-182-PEG3-K(palmitic acid)] not only showed improved stability but also increased affinity to the CD206 receptor through enhanced interaction with a hydrophobic binding motif of CD206. Peptide 1a showed superior in vitro activity in cell-based assays of macrophage activation which was restricted to CD206high M2-polarized macrophages. Improvement in responses was disproportionally skewed toward improved induction of phagocytosis including cancer cell phagocytosis. Peptide 1a reprogrammed the immune landscape in genetically engineered murine KPC pancreatic tumors toward increased innate immune surveillance and improved tumor control and effectively suppressed tumor growth of murine B16 melanoma allografts.
Collapse
Affiliation(s)
- Sitanshu S. Singh
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Raul Calvo
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Anju Kumari
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Rushikesh V. Sable
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Yuhong Fang
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Dingyin Tao
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Xin Hu
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Sarah Gray Castle
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Saifun Nahar
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Dandan Li
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Emily Major
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Tino W. Sanchez
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Rintaro Kato
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Xin Xu
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | | | - Liang Liu
- CPC Scientific Inc., San Jose, California
| | - Christopher A. LeClair
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Anton Simeonov
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Bolormaa Baljinnyam
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Mark J. Henderson
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Juan Marugan
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Udo Rudloff
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
4
|
Nielipińska D, Rubiak D, Pietrzyk-Brzezińska AJ, Małolepsza J, Błażewska KM, Gendaszewska-Darmach E. Stapled peptides as potential therapeutics for diabetes and other metabolic diseases. Biomed Pharmacother 2024; 180:117496. [PMID: 39362065 DOI: 10.1016/j.biopha.2024.117496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
The field of peptide drug research has experienced notable progress, with stapled peptides featuring stabilized α-helical conformation, emerging as a promising field. These peptides offer enhanced stability, cellular permeability, and binding affinity and exhibit potential in the treatment of diabetes and metabolic disorders. Stapled peptides, through the disruption of protein-protein interactions, present varied functionalities encompassing agonism, antagonism, and dual-agonism. This comprehensive review offers insight into the technology of peptide stapling and targeting of crucial molecular pathways associated with glucose metabolism, insulin secretion, and food intake. Additionally, we address the challenges in developing stapled peptides, including concerns pertaining to structural stability, peptide helicity, isomer mixture, and potential side effects.
Collapse
Affiliation(s)
- Dominika Nielipińska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland.
| | - Dominika Rubiak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland
| | - Agnieszka J Pietrzyk-Brzezińska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland
| | - Joanna Małolepsza
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland
| | - Katarzyna M Błażewska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland.
| | - Edyta Gendaszewska-Darmach
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland.
| |
Collapse
|
5
|
Li Y, Wu M, Fu Y, Xue J, Yuan F, Qu T, Rissanou AN, Wang Y, Li X, Hu H. Therapeutic stapled peptides: Efficacy and molecular targets. Pharmacol Res 2024; 203:107137. [PMID: 38522761 DOI: 10.1016/j.phrs.2024.107137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/26/2024]
Abstract
Peptide stapling, by employing a stable, preformed alpha-helical conformation, results in the production of peptides with improved membrane permeability and enhanced proteolytic stability, compared to the original peptides, and provides an effective solution to accelerate the rapid development of peptide drugs. Various reviews present peptide stapling chemistries, anchoring residues and one- or two-component cyclization, however, therapeutic stapled peptides have not been systematically summarized, especially focusing on various disease-related targets. This review highlights the latest advances in therapeutic peptide drug development facilitated by the application of stapling technology, including different stapling techniques, synthetic accessibility, applicability to biological targets, potential for solving biological problems, as well as the current status of development. Stapled peptides as therapeutic drug candidates have been classified and analysed mainly by receptor- and ligand-based stapled peptide design against various diseases, including cancer, infectious diseases, inflammation, and diabetes. This review is expected to provide a comprehensive reference for the rational design of stapled peptides for different diseases and targets to facilitate the development of therapeutic peptides with enhanced pharmacokinetic and biological properties.
Collapse
Affiliation(s)
- Yulei Li
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| | - Minghao Wu
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Yinxue Fu
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Jingwen Xue
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Fei Yuan
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Tianci Qu
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Anastassia N Rissanou
- Theoretical & Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - Yilin Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, 131 Dong'an Road, Shanghai 200032, China
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| | - Honggang Hu
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China.
| |
Collapse
|
6
|
Ran Y, Hu C, Wan J, Kang Q, Zhou R, Liu P, Ma D, Wang J, Tang L. Integrated investigation and experimental validation of PPARG as an oncogenic driver: implications for prognostic assessment and therapeutic targeting in hepatocellular carcinoma. Front Pharmacol 2023; 14:1298341. [PMID: 38044948 PMCID: PMC10690586 DOI: 10.3389/fphar.2023.1298341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/06/2023] [Indexed: 12/05/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARG), a key transcription factor involved in lipid metabolism and glucose homeostasis, has been implicated in various types of cancer. However, its precise role in cancer remains unclear. In this study, we conducted a comprehensive pan-cancer analysis of PPARG expression using various types of cancer obtained from public databases. We observed significant heterogeneity in PPARG expression across different types of cancer. The association between PPARG expression and patient prognosis was investigated using Cox proportional hazards regression models and survival analysis. Clinical features and protein expression levels in the cohort showed that PPARG expression was strongly associated, suggesting its potential as a therapeutic target. We also evaluated the prognostic potential of PPARG by analyzing immune infiltration and genomic stability. We experimentally validated the potential of PPARG as a therapeutic target by analyzing drug sensitivity profiles, molecular docking simulations, and in vitro cell proliferation assays associated with PPARG expression. We identified common expression patterns of PPARG with other genes involved in key carcinogenic pathways. This provides deeper insights into the molecular mechanisms underlying its carcinogenic role. Additionally, functional enrichment analysis revealed significant enrichment of genes related to drug metabolism, cell proliferation, and immune response pathways associated with PPARG. Our findings highlight the importance of PPARG in the broader biology of cancer and suggest its potential as a diagnostic and therapeutic target for specific types of cancer. The results of our study provide strong support for the potential role of PPARG as a promising prognostic biomarker and immunotherapeutic target across various types of cancer.
Collapse
Affiliation(s)
- Yunsheng Ran
- School of Pharmacy, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Chujiao Hu
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Junzhao Wan
- School of Pharmacy, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Qian Kang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ruixian Zhou
- Department of Acupuncture and Moxibustion, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ping Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Dan Ma
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jianta Wang
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Lei Tang
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
7
|
Sang P, Cai J. Unnatural helical peptidic foldamers as protein segment mimics. Chem Soc Rev 2023; 52:4843-4877. [PMID: 37401344 PMCID: PMC10389297 DOI: 10.1039/d2cs00395c] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Indexed: 07/05/2023]
Abstract
Unnatural helical peptidic foldamers have attracted considerable attention owing to their unique folding behaviours, diverse artificial protein binding mechanisms, and promising applications in chemical, biological, medical, and material fields. Unlike the conventional α-helix consisting of molecular entities of native α-amino acids, unnatural helical peptidic foldamers are generally comprised of well-defined backbone conformers with unique and unnatural structural parameters. Their folded structures usually arise from unnatural amino acids such as N-substituted glycine, N-substituted-β-alanine, β-amino acid, urea, thiourea, α-aminoxy acid, α-aminoisobutyric acid, aza-amino acid, aromatic amide, γ-amino acid, as well as sulfono-γ-AA amino acid. They can exhibit intriguing and predictable three-dimensional helical structures, generally featuring superior resistance to proteolytic degradation, enhanced bioavailability, and improved chemodiversity, and are promising in mimicking helical segments of various proteins. Although it is impossible to include every piece of research work, we attempt to highlight the research progress in the past 10 years in exploring unnatural peptidic foldamers as protein helical segment mimics, by giving some representative examples and discussing the current challenges and future perspectives. We expect that this review will help elucidate the principles of structural design and applications of existing unnatural helical peptidic foldamers in protein segment mimicry, thereby attracting more researchers to explore and generate novel unnatural peptidic foldamers with unique structural and functional properties, leading to more unprecedented and practical applications.
Collapse
Affiliation(s)
- Peng Sang
- Tianjian Laboratory of Advanced Biomedical Sciences, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
8
|
Kurtzhals P, Østergaard S, Nishimura E, Kjeldsen T. Derivatization with fatty acids in peptide and protein drug discovery. Nat Rev Drug Discov 2023; 22:59-80. [PMID: 36002588 DOI: 10.1038/s41573-022-00529-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 01/28/2023]
Abstract
Peptides and proteins are widely used to treat a range of medical conditions; however, they often have to be injected and their effects are short-lived. These shortcomings of the native structure can be addressed by molecular engineering, but this is a complex undertaking. A molecular engineering technology initially applied to insulin - and which has now been successfully applied to several biopharmaceuticals - entails the derivatization of peptides and proteins with fatty acids. Various protraction mechanisms are enabled by the specific characteristics and positions of the attached fatty acid. Furthermore, the technology can ensure a long half-life following oral administration of peptide drugs, can alter the distribution of peptides and may hold potential for tissue targeting. Due to the inherent safety and well-defined chemical nature of the fatty acids, this technology provides a versatile approach to peptide and protein drug discovery.
Collapse
|
9
|
α/Sulfono-γ-AA peptide hybrids agonist of GLP-1R with prolonged action both in vitro and in vivo. Acta Pharm Sin B 2022; 13:1648-1659. [PMID: 37139407 PMCID: PMC10149899 DOI: 10.1016/j.apsb.2022.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/22/2022] [Accepted: 10/14/2022] [Indexed: 11/01/2022] Open
Abstract
Peptides are increasingly important resources for biological and therapeutic development, however, their intrinsic susceptibility to proteolytic degradation represents a big hurdle. As a natural agonist for GLP-1R, glucagon-like peptide 1 (GLP-1) is of significant clinical interest for the treatment of type-2 diabetes mellitus, but its in vivo instability and short half-life have largely prevented its therapeutic application. Here, we describe the rational design of a series of α/sulfono-γ-AA peptide hybrid analogues of GLP-1 as the GLP-1R agonists. Certain GLP-1 hybrid analogues exhibited enhanced stability (t 1/2 > 14 days) compared to t 1/2 (<1 day) of GLP-1 in the blood plasma and in vivo. These newly developed peptide hybrids may be viable alternative of semaglutide for type-2 diabetes treatment. Additionally, our findings suggest that sulfono-γ-AA residues could be adopted to substitute canonical amino acids residues to improve the pharmacological activity of peptide-based drugs.
Collapse
|
10
|
Ali M, Namjoshi S, Benson HAE, Mohammed Y, Kumeria T. Dissolvable polymer microneedles for drug delivery and diagnostics. J Control Release 2022; 347:561-589. [PMID: 35525331 DOI: 10.1016/j.jconrel.2022.04.043] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
Abstract
Dissolvable transdermal microneedles (μND) are promising micro-devices used to transport a wide selection of active compounds into the skin. To provide an effective therapeutic outcome, μNDs must pierce the human stratum corneum (~10 to 20 μm), without rupturing or bending during penetration, then release their cargo at the predetermined area and time. The ability of dissolvable μND arrays/patches to sufficiently pierce the skin is a crucial requirement, which depends on the material composition, μND geometry and fabrication techniques. This comprehensive review not only provides contemporary knowledge on the μND design approaches, but also the materials science facilitating these delivery systems and the opportunities these advanced materials can provide to enhance clinical outcomes.
Collapse
Affiliation(s)
- Masood Ali
- Therapeutics Research Group, The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD 4102, Australia
| | - Sarika Namjoshi
- Therapeutics Research Group, The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD 4102, Australia; Vaxxas Pty Ltd, Brisbane, Woolloongabba, QLD 4102, Australia
| | - Heather A E Benson
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; Basil Hetzel institute for Translational Health Research, Adelaide, SA 5001, Australia.
| | - Yousuf Mohammed
- Therapeutics Research Group, The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD 4102, Australia.
| | - Tushar Kumeria
- School of Materials Science and Engineering, The University of New South Wales, Sydney. NSW 2052, Australia; Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW 2052, Australia; School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| |
Collapse
|
11
|
Yang Q, Zhou F, Tang X, Wang J, Feng H, Jiang W, Jin L, Jiang N, Yuan Y, Han J, Yan Z. Peptide-based long-acting co-agonists of GLP-1 and cholecystokinin 1 receptors as novel anti-diabesity agents. Eur J Med Chem 2022; 233:114214. [DOI: 10.1016/j.ejmech.2022.114214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/12/2022] [Accepted: 02/18/2022] [Indexed: 12/13/2022]
|
12
|
Bryce DA, Kitt JP, Harris JM. Raman Microscopy Investigation of GLP-1 Peptide Association with Supported Phospholipid Bilayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:14265-14274. [PMID: 34856805 DOI: 10.1021/acs.langmuir.1c01663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
A wide range of important biological processes occur at phospholipid membranes including cell signaling, where a peptide or small molecule targets a membrane-localized receptor protein. In this work, we report the adaptation of confocal Raman microscopy to quantify populations of unlabeled glucagon-like peptide-1 (GLP-1), a membrane-active 30-residue incretin peptide, in supported phospholipid bilayers deposited on the interior surfaces of wide-pore porous silica particles. Quantification of lipid bilayer-associated peptide is achieved by measuring the Raman scattering intensity of the peptide relative to that of the supported lipid bilayer, which serves as an internal standard. The dependence of the bilayer-associated GLP-1 population on the solution concentration of GLP-1 produces an isotherm used to determine the equilibrium constant for peptide-bilayer association and the maximum peptide surface coverage. The maximum coverage of GLP-1 in the lipid bilayer was found to be only 1/5th of a full monolayer based on its hydrodynamic radius. The saturation coverage, therefore, is not limited by the size of GLP-1 but by the ability of the bilayer to accommodate the peptide at high concentrations within the bilayer. Raman spectra show that GLP-1 association with the supported bilayer is accompanied by structural changes consistent with the intercalation of the peptide into the bilayer, where the observed increase in acyl-chain order would increase the lipid density and provide free volume needed to accommodate the peptide. These results were compared with previous measurements of the association of fluorescently labeled GLP-1 with a planar-supported bilayer; the unlabeled peptide exhibits a 3-fold greater affinity for the lipid bilayer on the porous silica support, suggesting that the fluorescent label alters the GLP-1 lipid bilayer association.
Collapse
Affiliation(s)
- David A Bryce
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112, United States
| | - Jay P Kitt
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112, United States
- Department of Biomedical Informatics, University of Utah, 421 Wakara Way Ste. 140, Salt Lake City, Utah 84108, United States
| | - Joel M Harris
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112, United States
| |
Collapse
|
13
|
Sang P, Zeng H, Lee C, Shi Y, Wang M, Pan C, Wei L, Huang C, Wu M, Shen W, Li X, Cai J. α/Sulfono-γ-AApeptide Hybrid Analogues of Glucagon with Enhanced Stability and Prolonged In Vivo Activity. J Med Chem 2021; 64:13893-13901. [PMID: 34506138 PMCID: PMC8903076 DOI: 10.1021/acs.jmedchem.1c01289] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Peptide drugs have the advantages of target specificity and good drugability and have become one of the most increasingly important hotspots in new drug research in biomedical sciences. However, peptide drugs generally have low bioavailability and metabolic stability, and therefore, the modification of existing peptide drugs for the purpose of improving stability and retaining activity is of viable importance. It is known that glucagon is an effective therapy for treating severe hypoglycemia, but its short half-life prevents its wide therapeutic use. Herein, we report that combined unnatural residues and long fatty acid conjugation afford potent α/sulfono-γ-AApeptide hybrid analogues of Glucagon with enhanced stability and prolonged in vivo activity. This strategy could be adopted to develop stabilized analogues of other short-acting bioactive peptides.
Collapse
Affiliation(s)
- Peng Sang
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Hongxiang Zeng
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Candy Lee
- Calibr at Scripps Research, 11119 N. Torrey Pines Rd., La Jolla, California 92037, United States
| | - Yan Shi
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Minghui Wang
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Cong Pan
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Lulu Wei
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Chenglong Huang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Mingjun Wu
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Weijun Shen
- Calibr at Scripps Research, 11119 N. Torrey Pines Rd., La Jolla, California 92037, United States
| | - Xi Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| |
Collapse
|
14
|
Wahid AA, Dunphy RW, Macpherson A, Gibson BG, Kulik L, Whale K, Back C, Hallam TM, Alkhawaja B, Martin RL, Meschede I, Laabei M, Lawson ADG, Holers VM, Watts AG, Crennell SJ, Harris CL, Marchbank KJ, van den Elsen JMH. Insights Into the Structure-Function Relationships of Dimeric C3d Fragments. Front Immunol 2021; 12:714055. [PMID: 34434196 PMCID: PMC8381054 DOI: 10.3389/fimmu.2021.714055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/21/2021] [Indexed: 11/13/2022] Open
Abstract
Cleavage of C3 to C3a and C3b plays a central role in the generation of complement-mediated defences. Although the thioester-mediated surface deposition of C3b has been well-studied, fluid phase dimers of C3 fragments remain largely unexplored. Here we show C3 cleavage results in the spontaneous formation of C3b dimers and present the first X-ray crystal structure of a disulphide-linked human C3d dimer. Binding studies reveal these dimers are capable of crosslinking complement receptor 2 and preliminary cell-based analyses suggest they could modulate B cell activation to influence tolerogenic pathways. Altogether, insights into the physiologically-relevant functions of C3d(g) dimers gained from our findings will pave the way to enhancing our understanding surrounding the importance of complement in the fluid phase and could inform the design of novel therapies for immune system disorders in the future.
Collapse
Affiliation(s)
- Ayla A. Wahid
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Rhys W. Dunphy
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Alex Macpherson
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
- UCB Pharma, Slough, United Kingdom
| | - Beth G. Gibson
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Liudmila Kulik
- Division of Rheumatology, University of Colorado, Aurora, CO, United States
| | | | - Catherine Back
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Thomas M. Hallam
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Bayan Alkhawaja
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Rebecca L. Martin
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | | | - Maisem Laabei
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | | | - V. Michael Holers
- Division of Rheumatology, University of Colorado, Aurora, CO, United States
| | - Andrew G. Watts
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
- Centre for Therapeutic Innovation, University of Bath, Bath, United Kingdom
| | - Susan J. Crennell
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Claire L. Harris
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Kevin J. Marchbank
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Jean M. H. van den Elsen
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
- Centre for Therapeutic Innovation, University of Bath, Bath, United Kingdom
| |
Collapse
|
15
|
Han C, Sun Y, Yang Q, Zhou F, Chen X, Wu L, Sun L, Han J. Stapled, Long-Acting Xenopus GLP-1-Based Dual GLP-1/Glucagon Receptor Agonists with Potent Therapeutic Efficacy for Metabolic Disease. Mol Pharm 2021; 18:2906-2923. [PMID: 34240881 DOI: 10.1021/acs.molpharmaceut.0c00995] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Novel peptidic glucagon receptor (GCGR) and glucagon-like peptide 1 receptor (GLP-1R) dual agonists are reported to have increased efficacy over GLP-1R monoagonists for the treatment of diabetes and obesity. We identified a novel Xenopus GLP-1-based dual GLP-1R/GCGR agonist (xGLP/GCG-13) designed with a proper activity ratio favoring the GLP-1R versus the GCGR. However, the clinical utility of xGLP/GCG-13 is limited by its short in vivo half-life. Starting from xGLP/GCG-13, dual Cys mutation was performed, followed by covalent side-chain stapling and serum albumin binder incorporation, resulting in a stabilized secondary structure, enhanced agonist potency at GLP-1R and GCGR, and improved stability. The lead peptide 2c (stapled xGLP/GCG-13 analogue with a palmitic acid albumin binder) exhibits balanced GLP-1R and GCGR activations and potent, long-lasting effects on in vivo glucose control. 2c was further explored pharmacologically in diet-induced obesity and db/db rodent models. Chronic administration of 2c potently induced body weight loss and hypoglycemic effects, improved glucose tolerance, increased energy expenditure, and normalized lipid metabolism and adiposity in relevant animal models. These results indicated that 2c has potential for development as a novel antidiabetic and/or antiobesity drug. Furthermore, we propose that the incorporation of a proper serum protein-binding motif into a di-Cys staple is an effective method for improving the stabilities and bioactivities of peptides. This approach is likely applicable to other therapeutic peptides, such as glucose-dependent insulin-tropic peptide receptor (GIPR) and GLP-1R dual agonists or GLP-1R/GCGR/GIPR triagonists.
Collapse
Affiliation(s)
- Chun Han
- Department of Chemistry, Changzhi University, Changzhi 046011, PR China
| | - Yuqing Sun
- Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, PR China
| | - Qimeng Yang
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Feng Zhou
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Xinyu Chen
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Lintao Wu
- Department of Chemistry, Changzhi University, Changzhi 046011, PR China
| | - Lidan Sun
- Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, PR China
| | - Jing Han
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| |
Collapse
|
16
|
Qian M, Zhang Q, Lu J, Zhang J, Wang Y, Shangguan W, Feng M, Feng J. Long-Acting Human Interleukin 2 Bioconjugate Modified with Fatty Acids by Sortase A. Bioconjug Chem 2021; 32:615-625. [PMID: 33656323 DOI: 10.1021/acs.bioconjchem.1c00062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human Interleukin 2 (IL-2) has already achieved impressive results as a therapeutic agent for cancer and autoimmune diseases. However, one of the limitations associated with the clinical application of IL-2 is its short half-life owing to rapid clearance by the kidneys. Modification with fatty acids, as an albumin noncovalent ligand with the advantage of deep penetration into tissues and high activity-to-mass ratio, is a commonly used approach to improve the half-life of native peptides and proteins. In this investigation, we attempted to extend the half-life of IL-2 through conjugation with a fatty acid using sortase A (srtA). We initially designed and optimized three IL-2 analogues with different peptide linkers between the C-terminus of IL-2 and srtA recognition sequence (LPETG). Among these, analogue A3 was validated as the optimal IL-2 analogue for further modification. Next, six fatty acid moieties with the same fatty acid and different hydrophilic spacers were conjugated to A3 through srtA. The six bioconjugates generated were screened for in vitro biological activity, among which bioconjugate B6 was identified as near-optimal to IL-2. Additionally, B6 could effectively bind albumin through the conjugated fatty acid, which contributed to a significant improvement in its pharmacokinetic properties in vivo. In summary, we have developed a novel IL-2 bioconjugate, B6, modified with fatty acids using srtA, which may effectively serve as a new-generation long-acting IL-2 immunotherapeutic agent.
Collapse
Affiliation(s)
- Mengxin Qian
- Department of Microbiological & Biochemical Pharmacy, School of Pharmacy, Fudan University, 201203 Shanghai, China.,State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Qingbin Zhang
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Jianguang Lu
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China.,Shanghai Duomirui Biotechnology Co., Ltd., 201203 Shanghai, China
| | - Jinhua Zhang
- Department of Microbiological & Biochemical Pharmacy, School of Pharmacy, Fudan University, 201203 Shanghai, China.,State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Yapeng Wang
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Wenwen Shangguan
- Department of Microbiological & Biochemical Pharmacy, School of Pharmacy, Fudan University, 201203 Shanghai, China.,State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Meiqing Feng
- Department of Microbiological & Biochemical Pharmacy, School of Pharmacy, Fudan University, 201203 Shanghai, China
| | - Jun Feng
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China.,Shanghai Duomirui Biotechnology Co., Ltd., 201203 Shanghai, China
| |
Collapse
|
17
|
Surface-engineered nanoliposomes with lipidated and non-lipidated peptide-dendrimeric scaffold for efficient transdermal delivery of a therapeutic agent: Development, characterization, toxicological and preclinical performance analyses. Eur J Pharm Biopharm 2020; 156:97-113. [DOI: 10.1016/j.ejpb.2020.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 11/23/2022]
|
18
|
Chen X, Fu J, Zhou F, Yang Q, Wang J, Feng H, Jiang W, Jin L, Tang X, Jiang N, Yin J, Han J. Stapled and Xenopus Glucagon-Like Peptide 1 (GLP-1)-Based Dual GLP-1/Gastrin Receptor Agonists with Improved Metabolic Benefits in Rodent Models of Obesity and Diabetes. J Med Chem 2020; 63:12595-12613. [PMID: 33125843 DOI: 10.1021/acs.jmedchem.0c00736] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Diabetes is characterized by pancreas dysfunction and is commonly associated with obesity. Hypoglycemic agents capable of improving β-cell function and reducing body weight therefore are gaining increasing interest. Though glucagon-like peptide 1 receptor (GLP-1R)/cholecystokinin 2 receptor (CCK-2R) dual agonist ZP3022 potently increases β-cell mass and improves glycemic control in diabetic db/db mice, the in vivo half-life (t1/2) is short, and its body weight reducing activity is limited. Here, we report the discovery of a series of novel GLP-1R/CCK-2R dual agonists. Starting from Xenopus GLP-1, dual cysteine mutation was conducted followed by covalent side chain stapling and albumin binder incorporation, resulting in a stabilized secondary structure, increased agonist potency, and improved stability. Further C-terminal conjugation of gastrin-6 generated GLP-1R/CCK-2R dual agonists, among which 6a and 6b showed higher stability and hypoglycemic activity than liraglutide and ZP3022. Desirably, 6a and 6b exhibited prominent metabolic benefits in diet-induced obesity mice without causing nausea responses and exerted considerable effects on β-cell restoration in db/db mice. These preclinical studies suggest the potential role of GLP-1R/CCK-2R dual agonists as effective agents for treating diabetes and related metabolic disorders.
Collapse
Affiliation(s)
- Xinyu Chen
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Junjie Fu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, PR China
| | - Feng Zhou
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Qimeng Yang
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Jialing Wang
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Hui Feng
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Wen Jiang
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Luofan Jin
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Xuelin Tang
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Neng Jiang
- Department of Pharmacy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, PR China
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, PR China
| | - Jing Han
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, PR China.,Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, PR China
| |
Collapse
|
19
|
Anananuchatkul T, Tsutsumi H, Miki T, Mihara H. hDM2 protein-binding peptides screened from stapled α-helical peptide phage display libraries with different types of staple linkers. Bioorg Med Chem Lett 2020; 30:127605. [PMID: 33038548 DOI: 10.1016/j.bmcl.2020.127605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/29/2020] [Accepted: 10/03/2020] [Indexed: 11/18/2022]
Abstract
Chemically modified peptide ligands were identified from α-helix peptide phage libraries with different types of staple linkers. The hDM2-protein was used as a representative target of protein-protein interactions to screen ligands for p53 binding sites in hDM2. Two types of staple linkers were used for the chemical modification of the peptide phage display libraries before affinity selection. The identified stapled peptides could bind to hDM2 competitively with the p53 peptide. The stapled peptide phage libraries developed in this study will improve the discovery of protein-protein interaction inhibitors through the synergistic effect of peptide units and staple linkers.
Collapse
Affiliation(s)
- Teerapat Anananuchatkul
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Hiroshi Tsutsumi
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Takayuki Miki
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Hisakazu Mihara
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan.
| |
Collapse
|
20
|
Lear S, Pflimlin E, Zhou Z, Huang D, Weng S, Nguyen-Tran V, Joseph SB, Roller S, Peterson S, Li J, Tremblay M, Schultz PG, Shen W. Engineering of a Potent, Long-Acting NPY2R Agonist for Combination with a GLP-1R Agonist as a Multi-Hormonal Treatment for Obesity. J Med Chem 2020; 63:9660-9671. [PMID: 32844654 DOI: 10.1021/acs.jmedchem.0c00740] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bariatric surgery results in increased intestinal secretion of hormones GLP-1 and anorexigenic PYY, which is believed to contribute to the clinical efficacy associated with the procedure. This observation raises the question whether combination treatment with gut hormone analogs might recapitulate the efficacy and mitigate the significant risks associated with surgery. Despite PYY demonstrating excellent efficacy and safety profiles with regard to food intake reduction, weight loss, and glucose control in preclinical animal models, PYY-based therapeutic development remains challenging given a low serum stability and half-life for the native peptide. Here, combined peptide stapling and PEG-fatty acid conjugation affords potent PYY analogs with >14 h rat half-lives, which are expected to translate into a human half-life suitable for once-weekly dosing. Excellent efficacy in glucose control, food intake reduction, and weight loss for lead candidate 22 in combination with our previously reported long-acting GLP-1 analog is demonstrated in a diet-induced obesity mouse model.
Collapse
Affiliation(s)
- Sam Lear
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Elsa Pflimlin
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Zhihong Zhou
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - David Huang
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Sharon Weng
- Intarcia Therapeutics, Inc., Research Triangle Park, 6 Davis Drive, Durham, North Carolina 27709, United States
| | - Van Nguyen-Tran
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Sean B Joseph
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Shane Roller
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Scott Peterson
- Intarcia Therapeutics, Inc., Research Triangle Park, 6 Davis Drive, Durham, North Carolina 27709, United States
| | - Jing Li
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Matthew Tremblay
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Peter G Schultz
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Weijun Shen
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| |
Collapse
|
21
|
Li X, Chen S, Zhang WD, Hu HG. Stapled Helical Peptides Bearing Different Anchoring Residues. Chem Rev 2020; 120:10079-10144. [DOI: 10.1021/acs.chemrev.0c00532] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Xiang Li
- School of Pharmacy, Second Military Medical University, Shanghai, China
- Insititute of Translational Medicine, Shanghai University, Shanghai, China
| | - Si Chen
- School of Medicine, Shanghai University, Shanghai, China
| | - Wei-Dong Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong-Gang Hu
- Insititute of Translational Medicine, Shanghai University, Shanghai, China
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| |
Collapse
|
22
|
Bird GH, Fu A, Escudero S, Godes M, Opoku-Nsiah K, Wales TE, Cameron MD, Engen JR, Danial NN, Walensky LD. Hydrocarbon-Stitched Peptide Agonists of Glucagon-Like Peptide-1 Receptor. ACS Chem Biol 2020; 15:1340-1348. [PMID: 32348108 DOI: 10.1021/acschembio.0c00308] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Glucagon-like peptide 1 (GLP-1) is a natural peptide agonist of the GLP-1 receptor (GLP-1R) found on pancreatic β-cells. Engagement of the receptor stimulates insulin release in a glucose-dependent fashion and increases β-cell mass, two ideal features for pharmacologic management of type 2 diabetes. Thus, intensive efforts have focused on developing GLP-1-based peptide agonists of GLP-1R for therapeutic application. A primary challenge has been the naturally short half-life of GLP-1 due to its rapid proteolytic degradation in vivo. Whereas mutagenesis and lipidation strategies have yielded clinical agents, we developed an alternative approach to preserving the structure and function of GLP-1 by all-hydrocarbon i, i + 7 stitching. This particular "stitch" is especially well-suited for reinforcing and protecting the structural fidelity of GLP-1. Lead constructs demonstrate striking proteolytic stability and potent biological activity in vivo. Thus, we report a facile approach to generating alternative GLP-1R agonists for glycemic control.
Collapse
Affiliation(s)
- Gregory H. Bird
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - Accalia Fu
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston Massachusetts 02215, United States
- Department of Cell Biology, Harvard Medical School, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Silvia Escudero
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - Marina Godes
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - Kwadwo Opoku-Nsiah
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - Thomas E. Wales
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Michael D. Cameron
- DMPK Core, Department of Molecular Medicine, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - John R. Engen
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Nika N. Danial
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston Massachusetts 02215, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Department of Medicine, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Loren D. Walensky
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
23
|
Tsao C, Zhang P, Yuan Z, Dong D, Wu K, Niu L, McMullen P, Luozhong S, Hung HC, Cheng YH, Jiang S. Zwitterionic Polymer Conjugated Glucagon-like Peptide-1 for Prolonged Glycemic Control. Bioconjug Chem 2020; 31:1812-1819. [DOI: 10.1021/acs.bioconjchem.0c00286] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Caroline Tsao
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Peng Zhang
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Zhefan Yuan
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Dianyu Dong
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
- School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering of Ministry of Education, Tianjin University, Tianjin, 300350, China
| | - Kan Wu
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Liqian Niu
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Patrick McMullen
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Sijin Luozhong
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Hsiang-Chieh Hung
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Yu-Hong Cheng
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Shaoyi Jiang
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
24
|
Lear S, Seo H, Lee C, Lei L, Amso Z, Huang D, Zou H, Zhou Z, Nguyen-Tran VTB, Shen W. Recombinant Expression and Stapling of a Novel Long-Acting GLP-1R Peptide Agonist. Molecules 2020; 25:molecules25112508. [PMID: 32481528 PMCID: PMC7321126 DOI: 10.3390/molecules25112508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/10/2020] [Accepted: 05/12/2020] [Indexed: 11/16/2022] Open
Abstract
Owing to their pleiotropic metabolic benefits, glucagon-like peptide-1 receptor (GLP-1R) agonists have been successfully utilized for treating metabolic diseases, such as type 2 diabetes and obesity. As part of our efforts in developing long-acting peptide therapeutics, we have previously reported a peptide engineering strategy that combines peptide side chain stapling with covalent integration of a serum protein-binding motif in a single step. Herein, we have used this strategy to develop a second generation extendin-4 analog rigidified with a symmetrical staple, which exhibits an excellent in vivo efficacy in an animal model of diabetes and obesity. To simplify the scale-up manufacturing of the lead GLP-1R agonist, a semisynthesis protocol was successfully developed, which involves recombinant expression of the linear peptide followed by attachment of a polyethylene glycol (PEG)-fatty acid staple in a subsequent chemical reaction step.
Collapse
|
25
|
Yang PY, Zou H, Amso Z, Lee C, Huang D, Woods AK, Nguyen-Tran VTB, Schultz PG, Shen W. New Generation Oxyntomodulin Peptides with Improved Pharmacokinetic Profiles Exhibit Weight Reducing and Anti-Steatotic Properties in Mice. Bioconjug Chem 2020; 31:1167-1176. [DOI: 10.1021/acs.bioconjchem.0c00093] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Peng-Yu Yang
- Calibr at The Scripps Research Institute, La Jolla, California 92037, United States
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Huafei Zou
- Calibr at The Scripps Research Institute, La Jolla, California 92037, United States
| | - Zaid Amso
- Calibr at The Scripps Research Institute, La Jolla, California 92037, United States
| | - Candy Lee
- Calibr at The Scripps Research Institute, La Jolla, California 92037, United States
| | - David Huang
- Calibr at The Scripps Research Institute, La Jolla, California 92037, United States
| | - Ashley K. Woods
- Calibr at The Scripps Research Institute, La Jolla, California 92037, United States
| | | | - Peter G. Schultz
- Calibr at The Scripps Research Institute, La Jolla, California 92037, United States
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Weijun Shen
- Calibr at The Scripps Research Institute, La Jolla, California 92037, United States
| |
Collapse
|
26
|
Stabilization and Transdermal Delivery of an Investigational Peptide Using MicroCor® Solid-State Dissolving Microstructure Arrays. J Pharm Sci 2020; 109:1288-1296. [DOI: 10.1016/j.xphs.2019.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023]
|
27
|
Pflimlin E, Zhou Z, Amso Z, Fu Q, Lee C, Muppiddi A, Joseph SB, Nguyen-Tran V, Shen W. Engineering a Potent, Long-Acting, and Periphery-Restricted Oxytocin Receptor Agonist with Anorexigenic and Body Weight Reducing Effects. J Med Chem 2019; 63:382-390. [PMID: 31850759 DOI: 10.1021/acs.jmedchem.9b01862] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The effects of oxytocin on food intake and body weight reduction have been demonstrated in both animal models and human clinical studies. Despite being efficacious, oxytocin is enzymatically unstable and thus considered to be unsuitable for long-term use in patients with obesity. Herein, a series of oxytocin derivatives were engineered through conjugation with fatty acid moieties that are known to exhibit high binding affinities to serum albumin. One analog (OT-12) in particular was shown to be a potent full agonist at the oxytocin receptor (OTR) in vitro with good selectivity and long half-life (24 h) in mice. Furthermore, OT-12 is peripherally restricted, with very limited brain exposure (1/190 of the plasma level). In a diet-induced obesity mouse model, daily subcutaneous administration of OT-12 exhibited more potent anorexigenic and body weight reducing effects than carbetocin. Thus, our results suggest that the long-acting, peripherally restricted OTR agonist may offer potential therapeutic benefits for obesity.
Collapse
Affiliation(s)
- Elsa Pflimlin
- Calibr at The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Zhihong Zhou
- Calibr at The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Zaid Amso
- Calibr at The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Qiangwei Fu
- Calibr at The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Candy Lee
- Calibr at The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Avinash Muppiddi
- Calibr at The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Sean B Joseph
- Calibr at The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Vân Nguyen-Tran
- Calibr at The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Weijun Shen
- Calibr at The Scripps Research Institute , La Jolla , California 92037 , United States
| |
Collapse
|
28
|
Singh P, Carrier A, Chen Y, Lin S, Wang J, Cui S, Zhang X. Polymeric microneedles for controlled transdermal drug delivery. J Control Release 2019; 315:97-113. [DOI: 10.1016/j.jconrel.2019.10.022] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/09/2019] [Accepted: 10/12/2019] [Indexed: 01/03/2023]
|
29
|
Zainal-Abidin MH, Hayyan M, Ngoh GC, Wong WF, Looi CY. Emerging frontiers of deep eutectic solvents in drug discovery and drug delivery systems. J Control Release 2019; 316:168-195. [DOI: 10.1016/j.jconrel.2019.09.019] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 01/02/2023]
|
30
|
Rajoli RKR, Flexner C, Chiong J, Owen A, Donnelly RF, Larrañeta E, Siccardi M. Modelling the intradermal delivery of microneedle array patches for long-acting antiretrovirals using PBPK. Eur J Pharm Biopharm 2019; 144:101-109. [PMID: 31525446 DOI: 10.1016/j.ejpb.2019.09.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/01/2019] [Accepted: 09/12/2019] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Existing HIV therapy using oral antiretrovirals (ARVs) can result in pill fatigue and sub-optimal adherence. Microneedle array patches (MAPs) offer non-invasive, blood-free and painless drug delivery, and may improve patient adherence. The objective of this study was to develop a novel physiologically-based pharmacokinetic (PBPK) model to simulate the systemic pharmacokinetics of cabotegravir and rilpivirine MAPs using the intradermal route. METHODS The developed PBPK models were qualified against observed pharmacokinetic data after intramuscular (IM) and intradermal administration of long-acting nanoformulated rilpivirine to rats, and for IM administration of both drugs to healthy adults. Qualified models were then utilised to estimate suitable MAP characteristics (e.g. nanoformulation dose and release rates) and inform dosing strategies to maintain plasma concentrations above target trough concentrations for the designated dosing interval. RESULTS PBPK models simulated q4-weekly loading and maintenance doses of 360 mg and 180 mg for long-acting formulated cabotegravir between the release rates of 1 × 10-3-3 × 10-3h-1 and 1 × 10-3-1.5 × 10-3h-1 respectively, for a 70 kg adult. Estimated patch size was 60 cm2 for a 360 mg dose of cabotegravir. For q4-weekly dosing, rilpivirine required a 1080 mg loading dose and a 540 mg maintenance dose with release rates of 1.5 × 10-3-2.5 × 10-3h-1 and 5 × 10-4-1 × 10-3h-1, respectively. Weekly dosing was also evaluated to assess the potential application from a smaller patch size. The ability to self-administer via a patch that is only left in place for a short duration makes longer durations less important than for some other long-acting approaches. Weekly cabotegravir required 60 mg between release rates 7 × 10-3-9 × 10-3h-1 and rilpivirine required 270 mg and 180 mg respectively between release rates of 7 × 10-3-9 × 10-3h-1. DISCUSSION This model estimated optimal dose and release rates for cabotegravir and rilpivirine MAPs. Our approach provides a computational platform to support rational development of intradermal administration strategies to tackle problems associated with chronic oral ARV administration.
Collapse
Affiliation(s)
- Rajith K R Rajoli
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK.
| | - Charles Flexner
- Johns Hopkins University School of Medicine and Bloomberg School of Public Health, Baltimore, MD, USA
| | - Justin Chiong
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Andrew Owen
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast, UK
| | - Eneko Larrañeta
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast, UK
| | - Marco Siccardi
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
31
|
Pflimlin E, Lear S, Lee C, Yu S, Zou H, To A, Joseph S, Nguyen-Tran V, Tremblay MS, Shen W. Design of a Long-Acting and Selective MEG-Fatty Acid Stapled Prolactin-Releasing Peptide Analog. ACS Med Chem Lett 2019; 10:1166-1172. [PMID: 31413801 DOI: 10.1021/acsmedchemlett.9b00182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/05/2019] [Indexed: 01/12/2023] Open
Abstract
Anorexigenic peptides offer promise as potential therapies targeting the escalating global obesity epidemic. Prolactin-releasing peptide (PrRP), a novel member of the RFamide family secreted by the hypothalamus, shows therapeutic potential by decreasing food intake and body weight in rodent models via GPR10 activation. Here we describe the design of a long-acting PrRP using our recently developed novel multiple ethylene glycol-fatty acid (MEG-FA) stapling platform. By incorporating serum albumin binding fatty acids onto a covalent side chain staple, we have generated a series of MEG-FA stapled PrRP analogs with enhanced serum stability and in vivo half-life. Our lead compound 18-S4 exhibits good in vitro potency and selectivity against GPR10, improved serum stability, and extended in vivo half-life (7.8 h) in mouse. Furthermore, 18-S4 demonstrates a potent body weight reduction effect in a diet-induced obesity (DIO) mouse model, representing a promising long-acting PrRP analog for further evaluation in the chronic obesity setting.
Collapse
Affiliation(s)
- Elsa Pflimlin
- Calibr at Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Sam Lear
- Calibr at Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Candy Lee
- Calibr at Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Shan Yu
- Calibr at Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Huafei Zou
- Calibr at Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Andrew To
- Calibr at Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Sean Joseph
- Calibr at Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Van Nguyen-Tran
- Calibr at Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Matthew S. Tremblay
- Calibr at Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Weijun Shen
- Calibr at Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| |
Collapse
|
32
|
Engineering PEG-fatty acid stapled, long-acting peptide agonists for G protein-coupled receptors. Methods Enzymol 2019; 622:183-200. [DOI: 10.1016/bs.mie.2019.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
33
|
Fu C, Chen Q, Zheng F, Yang L, Li H, Zhao Q, Wang X, Wang L, Wang Q. Genetically Encoding a Lipidated Amino Acid for Extension of Protein Half-Life in vivo. Angew Chem Int Ed Engl 2018; 58:1392-1396. [DOI: 10.1002/anie.201811837] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/15/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Caiyun Fu
- College of Life Sciences; Zhejiang Sci-Tech University; Hangzhou 310018 China
| | - Qi Chen
- College of Life Sciences; Zhejiang Sci-Tech University; Hangzhou 310018 China
- Hangzhou Research Institute of Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Hangzhou 310018 China
| | - Feng Zheng
- Hangzhou Research Institute of Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Hangzhou 310018 China
| | - Liu Yang
- Hangzhou Research Institute of Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Hangzhou 310018 China
| | - Haorong Li
- Hangzhou Research Institute of Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Hangzhou 310018 China
| | - Qianqian Zhao
- BGI Education Center; University of Chinese Academy of Sciences; Shenzhen 518083 China
| | - Xiumei Wang
- Jiuyuan Gene Engineering Co. LTD; Hangzhou 310018 China
| | - Lei Wang
- Department of Pharmaceutical Chemistry; University of California San Francisco; San Francisco CA 94158 USA
| | - Qian Wang
- Hangzhou Research Institute of Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Hangzhou 310018 China
| |
Collapse
|
34
|
Fu C, Chen Q, Zheng F, Yang L, Li H, Zhao Q, Wang X, Wang L, Wang Q. Genetically Encoding a Lipidated Amino Acid for Extension of Protein Half-Life in vivo. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201811837] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Caiyun Fu
- College of Life Sciences; Zhejiang Sci-Tech University; Hangzhou 310018 China
| | - Qi Chen
- College of Life Sciences; Zhejiang Sci-Tech University; Hangzhou 310018 China
- Hangzhou Research Institute of Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Hangzhou 310018 China
| | - Feng Zheng
- Hangzhou Research Institute of Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Hangzhou 310018 China
| | - Liu Yang
- Hangzhou Research Institute of Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Hangzhou 310018 China
| | - Haorong Li
- Hangzhou Research Institute of Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Hangzhou 310018 China
| | - Qianqian Zhao
- BGI Education Center; University of Chinese Academy of Sciences; Shenzhen 518083 China
| | - Xiumei Wang
- Jiuyuan Gene Engineering Co. LTD; Hangzhou 310018 China
| | - Lei Wang
- Department of Pharmaceutical Chemistry; University of California San Francisco; San Francisco CA 94158 USA
| | - Qian Wang
- Hangzhou Research Institute of Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Hangzhou 310018 China
| |
Collapse
|
35
|
Tian Y, Zou H, An P, Zhou Z, Shen W, Lin Q. Design of Stapled Oxyntomodulin Analogs Containing Functionalized Biphenyl Cross-Linkers. Tetrahedron 2018; 75:286-295. [PMID: 30581241 DOI: 10.1016/j.tet.2018.11.060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A panel of three lipid-modified, functionalized biphenyl cross-linkers (fBph) were synthesized and subsequently employed in the preparation of the stapled oxyntomodulin (OXM) analogs. In a luciferase-based reporter assay, these stapled OXM analogs showed varying degree of potency in activating GLP-1R and GCGR, presumably due to the disparate effect of the lipid chains on the local environment close to the ligand-receptor binding interface. In particular, the fBph-1 cross-linked peptide with the lipid chain attached to position-3 of the biphenyl cross-linker exhibited the highest dual agonist activity.
Collapse
Affiliation(s)
- Yulin Tian
- Department of Chemistry, State University of New York at Buffalo, Buffalo, New York 14260, United States.,Transira Therapeutics, 1576 Sweet Home Road, Baird Research Park, Amherst, New York 14228, United States
| | - Huafei Zou
- California Institute for Biomedical Research (Calibr), 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Peng An
- Department of Chemistry, State University of New York at Buffalo, Buffalo, New York 14260, United States.,Transira Therapeutics, 1576 Sweet Home Road, Baird Research Park, Amherst, New York 14228, United States
| | - Zhihong Zhou
- California Institute for Biomedical Research (Calibr), 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Weijun Shen
- California Institute for Biomedical Research (Calibr), 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Qing Lin
- Department of Chemistry, State University of New York at Buffalo, Buffalo, New York 14260, United States.,Transira Therapeutics, 1576 Sweet Home Road, Baird Research Park, Amherst, New York 14228, United States
| |
Collapse
|
36
|
Moonschi FH, Hughes CB, Mussman GM, Fowlkes JL, Richards CI, Popescu I. Advances in micro- and nanotechnologies for the GLP-1-based therapy and imaging of pancreatic beta-cells. Acta Diabetol 2018; 55:405-418. [PMID: 29264724 DOI: 10.1007/s00592-017-1086-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/03/2017] [Indexed: 12/20/2022]
Abstract
Therapies to prevent diabetes in particular the progressive loss of β-cell mass and function and/or to improve the dysregulated metabolism associated with diabetes are highly sought. The incretin-based therapy comprising GLP-1R agonists and DPP-4 inhibitors have represented a major focus of pharmaceutical R&D over the last decade. The incretin hormone GLP-1 has powerful antihyperglycemic effect through direct stimulation of insulin biosynthesis and secretion within the β-cells; it normalizes β-cell sensitivity to glucose, has an antiapoptotic role, stimulates β-cell proliferation and differentiation, and inhibits glucagon secretion. However, native GLP-1 therapy is inappropriate due to the rapid post-secretory inactivation by DPP-4. Therefore, incretin mimetics developed on the backbone of the GLP-1 or exendin-4 molecule have been developed to behave as GLP-1R agonists but to display improved stability and clinical efficacy. New formulations of incretins and their analogs based on micro- and nanomaterials (i.e., PEG, PLGA, chitosan, liposomes and silica) and innovative encapsulation strategies have emerged to achieve a better stability of the incretin, to improve its pharmacokinetic profile, to lower the administration frequency or to allow another administration route and to display fewer adverse effects. An important advantage of these formulations is that they can also be used at the targeted non-invasive imaging of the beta-cell mass. This review therefore focuses on the current state of these efforts as the next step in the therapeutic evolution of this class of antidiabetic drugs.
Collapse
Affiliation(s)
- Faruk H Moonschi
- Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Corey B Hughes
- Barnstable Brown Kentucky Diabetes Center, University of Kentucky, 900 S. Limestone, CTW 469, Lexington, KY, 40536, USA
| | - George M Mussman
- Barnstable Brown Kentucky Diabetes Center, University of Kentucky, 900 S. Limestone, CTW 469, Lexington, KY, 40536, USA
| | - John L Fowlkes
- Barnstable Brown Kentucky Diabetes Center, University of Kentucky, 900 S. Limestone, CTW 469, Lexington, KY, 40536, USA
| | - Chris I Richards
- Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Iuliana Popescu
- Barnstable Brown Kentucky Diabetes Center, University of Kentucky, 900 S. Limestone, CTW 469, Lexington, KY, 40536, USA.
| |
Collapse
|
37
|
Yang PY, Zou H, Lee C, Muppidi A, Chao E, Fu Q, Luo X, Wang D, Schultz PG, Shen W. Stapled, Long-Acting Glucagon-like Peptide 2 Analog with Efficacy in Dextran Sodium Sulfate Induced Mouse Colitis Models. J Med Chem 2018. [PMID: 29528634 DOI: 10.1021/acs.jmedchem.7b00768] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glucagon-like peptide 2 (GLP-2) is a hormone that has been shown to stimulate intestinal growth and attenuate intestinal inflammation. Despite being efficacious in a variety of animal models of disease, its therapeutic potential is hampered by the short half-life in vivo. We now describe a highly potent, stapled long-acting GLP-2 analog, peptide 10, that has a more than 10-fold longer half-life than teduglutide and improved intestinotrophic and anti-inflammatory effects in mouse models of DSS-induced colitis.
Collapse
Affiliation(s)
- Peng-Yu Yang
- California Institute for Biomedical Research , La Jolla , California 92037 , United States.,Department of Chemistry, The Skaggs Institute for Chemical Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Huafei Zou
- California Institute for Biomedical Research , La Jolla , California 92037 , United States
| | - Candy Lee
- California Institute for Biomedical Research , La Jolla , California 92037 , United States
| | - Avinash Muppidi
- California Institute for Biomedical Research , La Jolla , California 92037 , United States
| | - Elizabeth Chao
- California Institute for Biomedical Research , La Jolla , California 92037 , United States
| | - Qiangwei Fu
- California Institute for Biomedical Research , La Jolla , California 92037 , United States
| | - Xiaozhou Luo
- Department of Chemistry, The Skaggs Institute for Chemical Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Danling Wang
- California Institute for Biomedical Research , La Jolla , California 92037 , United States
| | - Peter G Schultz
- California Institute for Biomedical Research , La Jolla , California 92037 , United States.,Department of Chemistry, The Skaggs Institute for Chemical Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Weijun Shen
- California Institute for Biomedical Research , La Jolla , California 92037 , United States
| |
Collapse
|
38
|
Ye Y, Yu J, Wen D, Kahkoska AR, Gu Z. Polymeric microneedles for transdermal protein delivery. Adv Drug Deliv Rev 2018; 127:106-118. [PMID: 29408182 PMCID: PMC6020694 DOI: 10.1016/j.addr.2018.01.015] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 12/24/2017] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
The intrinsic properties of therapeutic proteins generally present a major impediment for transdermal delivery, including their relatively large molecule size and susceptibility to degradation. One solution is to utilize microneedles (MNs), which are capable of painlessly traversing the stratum corneum and directly translocating protein drugs into the systematic circulation. MNs can be designed to incorporate appropriate structural materials as well as therapeutics or formulations with tailored physicochemical properties. This platform technique has been applied to deliver drugs both locally and systemically in applications ranging from vaccination to diabetes and cancer therapy. This review surveys the current design and use of polymeric MNs for transdermal protein delivery. The clinical potential and future translation of MNs are also discussed.
Collapse
Affiliation(s)
- Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jicheng Yu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Di Wen
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Anna R Kahkoska
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
39
|
Marya, Khan H, Nabavi SM, Habtemariam S. Anti-diabetic potential of peptides: Future prospects as therapeutic agents. Life Sci 2017; 193:153-158. [PMID: 29055800 DOI: 10.1016/j.lfs.2017.10.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a metabolic disorder in which the glucose level in blood exceeds beyond the normal level. Persistent hyperglycemia leads to diabetes late complication and obviously account for a large number of morbidity and mortality worldwide. Numerous therapeutic options are available for the treatment of diabetes including insulin for type I and oral tablets for type II, but its effective management is still a dream. To date, several options are under investigation in various research laboratories for efficacious and safer agents. Of them, peptides are currently amongst the most widely investigated potential therapeutic agents whose design and optimal uses are under development. A number of natural and synthetic peptides have so far been found with outstanding antidiabetic effect mediated through diverse mechanisms. The applications of new emerging techniques and drug delivery systems further offer opportunities to achieve the desired target outcomes. Some outstanding peptides in preclinical and clinical studies with better efficacy and safety profile have already been identified. Further detail studies on these peptides may therefore lead to significant clinically useful antidiabetic agents.
Collapse
Affiliation(s)
- Marya
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan.
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories & Herbal Analysis Services, University of Greenwich, Central Avenue, Charham-Maritime, Kent ME4 4TB, UK
| |
Collapse
|
40
|
Wu Y, Villa F, Maman J, Lau YH, Dobnikar L, Simon AC, Labib K, Spring DR, Pellegrini L. Targeting the Genome-Stability Hub Ctf4 by Stapled-Peptide Design. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201705611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yuteng Wu
- Department of Chemistry; University of Cambridge; Cambridge CB2 1EW UK
| | - Fabrizio Villa
- MRC protein phosphorylation and ubiquitylation unit; University of Dundee; Dundee DD1 5EH UK
| | - Joseph Maman
- Department of Biochemistry; University of Cambridge; Cambridge CB2 1GA UK
| | - Yu Heng Lau
- Department of Chemistry; University of Cambridge; Cambridge CB2 1EW UK
- Current address: School of Chemistry; The University of Sydney (Australia)
| | - Lina Dobnikar
- Department of Chemistry; University of Cambridge; Cambridge CB2 1EW UK
| | - Aline C. Simon
- Department of Biochemistry; University of Cambridge; Cambridge CB2 1GA UK
| | - Karim Labib
- MRC protein phosphorylation and ubiquitylation unit; University of Dundee; Dundee DD1 5EH UK
| | - David R. Spring
- Department of Chemistry; University of Cambridge; Cambridge CB2 1EW UK
| | - Luca Pellegrini
- Department of Biochemistry; University of Cambridge; Cambridge CB2 1GA UK
| |
Collapse
|
41
|
Wu Y, Villa F, Maman J, Lau YH, Dobnikar L, Simon AC, Labib K, Spring DR, Pellegrini L. Targeting the Genome-Stability Hub Ctf4 by Stapled-Peptide Design. Angew Chem Int Ed Engl 2017; 56:12866-12872. [PMID: 28815832 DOI: 10.1002/anie.201705611] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/26/2017] [Indexed: 12/26/2022]
Abstract
The exploitation of synthetic lethality by small-molecule targeting of pathways that maintain genomic stability is an attractive chemotherapeutic approach. The Ctf4/AND-1 protein hub, which links DNA replication, repair, and chromosome segregation, represents a novel target for the synthetic lethality approach. Herein, we report the design, optimization, and validation of double-click stapled peptides encoding the Ctf4-interacting peptide (CIP) of the replicative helicase subunit Sld5. By screening stapling positions in the Sld5 CIP, we identified an unorthodox i,i+6 stapled peptide with improved, submicromolar binding to Ctf4. The mode of interaction with Ctf4 was confirmed by a crystal structure of the stapled Sld5 peptide bound to Ctf4. The stapled Sld5 peptide was able to displace the Ctf4 partner DNA polymerase α from the replisome in yeast extracts. Our study provides proof-of-principle evidence for the development of small-molecule inhibitors of the human CTF4 orthologue AND-1.
Collapse
Affiliation(s)
- Yuteng Wu
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Fabrizio Villa
- MRC protein phosphorylation and ubiquitylation unit, University of Dundee, Dundee, DD1 5EH, UK
| | - Joseph Maman
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK
| | - Yu Heng Lau
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.,Current address: School of Chemistry, The University of Sydney (Australia)
| | - Lina Dobnikar
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Aline C Simon
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK
| | - Karim Labib
- MRC protein phosphorylation and ubiquitylation unit, University of Dundee, Dundee, DD1 5EH, UK
| | - David R Spring
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Luca Pellegrini
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK
| |
Collapse
|
42
|
Banerjee A, Ibsen K, Iwao Y, Zakrewsky M, Mitragotri S. Transdermal Protein Delivery Using Choline and Geranate (CAGE) Deep Eutectic Solvent. Adv Healthc Mater 2017; 6. [PMID: 28337858 DOI: 10.1002/adhm.201601411] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 02/21/2017] [Indexed: 12/20/2022]
Abstract
Transdermal delivery of peptides and other biological macromolecules is limited due to skin's inherent low permeability. Here, the authors report the use of a deep eutectic solvent, choline and geranate (CAGE), to enhance topical delivery of proteins such as bovine serum albumin (BSA, molecular weight: ≈66 kDa), ovalbumin (OVA, molecular weight: ≈45 kDa) and insulin (INS, molecular weight: 5.8 kDa). CAGE enhances permeation of BSA, OVA, and insulin into porcine skin ex vivo, penetrating deep into the epidermis and dermis. Studies using tritium-labeled BSA and fluorescein isothiocyanate labeled insulin show significantly enhanced delivery of proteins into and across porcine skin, penetrating the skin in a time-dependent manner. Fourier transform IR spectra of porcine stratum corneum (SC) samples before and after incubation in CAGE show a reduction in peak area attributed to SC lipid content, suggesting lipid extraction from the SC. Circular dichroism confirms that CAGE does not affect insulin's secondary conformation. In vivo studies in rats show that topical application of 10 U insulin dispersed in CAGE (25 U kg-1 insulin dose) leads to a highly significant 40% drop in blood glucose levels in 4 h that is relatively sustained for 12 h. Taken together, these studies demonstrate that CAGE is a promising vehicle for transdermal delivery of therapeutic proteins; specifically, as a noninvasive delivery alternative to injectable insulin for the treatment of diabetes.
Collapse
Affiliation(s)
- Amrita Banerjee
- Department of Chemical Engineering and Center for Bioengineering University of California Santa Barbara Santa Barbara CA 93106 USA
| | - Kelly Ibsen
- Department of Chemical Engineering and Center for Bioengineering University of California Santa Barbara Santa Barbara CA 93106 USA
| | - Yasunori Iwao
- Department of Pharmaceutical Engineering School of Pharmaceutical Sciences University of Shizuoka Shizuoka 422‐8526 Japan
| | - Michael Zakrewsky
- Department of Chemical Engineering and Center for Bioengineering University of California Santa Barbara Santa Barbara CA 93106 USA
| | - Samir Mitragotri
- Department of Chemical Engineering and Center for Bioengineering University of California Santa Barbara Santa Barbara CA 93106 USA
| |
Collapse
|
43
|
Boettcher BR. Gastric bypass surgery mimetic approaches. Drug Discov Today 2017; 22:1242-1249. [PMID: 28576430 DOI: 10.1016/j.drudis.2017.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/11/2017] [Accepted: 04/18/2017] [Indexed: 01/10/2023]
Abstract
Gastric bypass surgery is effectively a polypharmacological approach for treatment of obesity, type 2 diabetes and nonalcoholic steatohepatitis (NASH). The gastric bypass mimetic approaches reviewed are fixed-dose combinatorial pharmacological approaches. There are two key concepts incorporated into these gastric bypass surgery mimetic approaches. The first key concept is that the combination of glucagon-like peptide 1 (GLP-1) and fibroblast growth factor 21 (FGF21) is essential for success of any gastric bypass surgery mimetic approach. This combination affords the potential for durable weight loss, glycemic control and reduction in liver lipids. The second key concept is that a fixed-dose combination approach is preferred over post-approval combination of the individual components because the individual components alone often lack sufficient efficacy for development.
Collapse
|
44
|
Wang Y, Bruno BJ, Cornillie S, Nogieira JM, Chen D, Cheatham TE, Lim CS, Chou DHC. Application of Thiol-yne/Thiol-ene Reactions for Peptide and Protein Macrocyclizations. Chemistry 2017; 23:7087-7092. [DOI: 10.1002/chem.201700572] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Yuanxiang Wang
- Department of Biochemistry; University of Utah; 15 N, Medical Drive East 4100 Salt Lake City UT 84112 USA
| | - Benjamin J. Bruno
- Department of Pharmaceutics and Pharmaceutical Chemistry; University of Utah; 30 S 2000 E, Rm 2916 Salt Lake City UT 84112 USA
| | - Sean Cornillie
- Department of Medicinal Chemistry; University of Utah; 30 S 2000 E, Rm 4914 Salt Lake City UT 84112 USA
| | - Jason M. Nogieira
- Department of Biochemistry; University of Utah; 15 N, Medical Drive East 4100 Salt Lake City UT 84112 USA
| | - Diao Chen
- Department of Biochemistry; University of Utah; 15 N, Medical Drive East 4100 Salt Lake City UT 84112 USA
| | - Thomas E. Cheatham
- Department of Medicinal Chemistry; University of Utah; 30 S 2000 E, Rm 4914 Salt Lake City UT 84112 USA
| | - Carol S. Lim
- Department of Pharmaceutics and Pharmaceutical Chemistry; University of Utah; 30 S 2000 E, Rm 2916 Salt Lake City UT 84112 USA
| | - Danny Hung-Chieh Chou
- Department of Biochemistry; University of Utah; 15 N, Medical Drive East 4100 Salt Lake City UT 84112 USA
| |
Collapse
|
45
|
Wang M, Hu L, Xu C. Recent advances in the design of polymeric microneedles for transdermal drug delivery and biosensing. LAB ON A CHIP 2017; 17:1373-1387. [PMID: 28352876 DOI: 10.1039/c7lc00016b] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Microneedles are an efficient and minimally invasive approach to transdermal drug delivery and extraction of skin interstitial fluid. Compared to solid microneedles made of silicon, metals and ceramics, polymeric microneedles have attracted extensive attention due to their excellent biocompatibility, biodegradability and nontoxicity. They are easy to fabricate in large scale and can load drugs in high amounts. More importantly, polymers with different degradation profiles, swelling properties, and responses to biological/physical stimuli can be employed to fabricate polymeric microneedles with different mechanical properties and performance. This review provides a guideline for the selection of polymers and the corresponding fabrication methods for polymeric microneedles while summarizing their recent application in drug delivery and fluid extraction. It should be noted that although polymeric microneedles can achieve efficient transdermal delivery of drugs, their wide applications were limited by their unsatisfactory transdermal therapeutic efficiency. Delivery of nanomedicines that incorporate drugs into functional nanoparticles/capsules can address this problem and thus may be an interesting direction in the future.
Collapse
Affiliation(s)
- Min Wang
- School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| | | | | |
Collapse
|
46
|
Xuan W, Shao S, Schultz PG. Protein Crosslinking by Genetically Encoded Noncanonical Amino Acids with Reactive Aryl Carbamate Side Chains. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201611841] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Weimin Xuan
- Department of Chemistry; the Scripps Research Institute; 10550 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Sida Shao
- Department of Chemistry; the Scripps Research Institute; 10550 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Peter G. Schultz
- Department of Chemistry; the Scripps Research Institute; 10550 N. Torrey Pines Road La Jolla CA 92037 USA
| |
Collapse
|
47
|
Xuan W, Shao S, Schultz PG. Protein Crosslinking by Genetically Encoded Noncanonical Amino Acids with Reactive Aryl Carbamate Side Chains. Angew Chem Int Ed Engl 2017; 56:5096-5100. [PMID: 28371162 DOI: 10.1002/anie.201611841] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/06/2017] [Indexed: 01/08/2023]
Abstract
The use of genetically encoded noncanonical amino acids (ncAAs) to construct crosslinks within or between proteins has emerged as a useful method to enhance protein stability, investigate protein-protein interactions, and improve the pharmacological properties of proteins. We report ncAAs with aryl carbamate side chains (PheK and FPheK) that can react with proximal nucleophilic residues to form intra- or intermolecular protein crosslinks. We evolved a pyrrolysyl-tRNA synthetase that incorporates site-specifically PheK and FPheK into proteins in both E. coli and mammalian cells. PheK and FPheK when incorporated into proteins showed good stability during protein expression and purification. FPheK reacted with adjacent Lys, Cys, and Tyr residues in thioredoxin in high yields. In addition, crosslinks could be formed between FPheK and Lys residue of two interacting proteins, including the heavy chain and light chain of an antibody Fab.
Collapse
Affiliation(s)
- Weimin Xuan
- Department of Chemistry, the Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Sida Shao
- Department of Chemistry, the Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Peter G Schultz
- Department of Chemistry, the Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|
48
|
Huang C, Wille CB, He H, Reddy VBG, Nargund RP, Lin S, Palani A. Late-stage lipidation of fully elaborated tryptophan-containing peptides for improved pharmacokinetics. Tetrahedron Lett 2017. [DOI: 10.1016/j.tetlet.2017.02.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
49
|
Wang Y, Du J, Zou H, Liu Y, Zhang Y, Gonzalez J, Chao E, Lu L, Yang P, Parker H, Nguyen-Tran V, Shen W, Wang D, Schultz PG, Wang F. Multifunctional Antibody Agonists Targeting Glucagon-like Peptide-1, Glucagon, and Glucose-Dependent Insulinotropic Polypeptide Receptors. Angew Chem Int Ed Engl 2016; 55:12475-8. [PMID: 27595986 DOI: 10.1002/anie.201606321] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/05/2016] [Indexed: 01/04/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) receptor (GLP-1R), glucagon (GCG) receptor (GCGR), and glucose-dependent insulinotropic polypeptide (GIP, also known as gastric inhibitory polypeptide) receptor (GIPR), are three metabolically related peptide hormone receptors. A novel approach to the generation of multifunctional antibody agonists that activate these receptors has been developed. Native or engineered peptide agonists for GLP-1R, GCGR, and GIPR were fused to the N-terminus of the heavy chain or light chain of an antibody, either alone or in pairwise combinations. The fusion proteins have similar in vitro biological activities on the cognate receptors as the corresponding peptides, but circa 100-fold longer plasma half-lives. The GLP-1R mono agonist and GLP-1R/GCGR dual agonist antibodies both exhibit potent effects on glucose control and body weight reduction in mice, with the dual agonist antibody showing enhanced activity in the latter.
Collapse
Affiliation(s)
- Ying Wang
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Jintang Du
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Huafei Zou
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Yan Liu
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Yuhan Zhang
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Jose Gonzalez
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Elizabeth Chao
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Lucy Lu
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Pengyu Yang
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Holly Parker
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Van Nguyen-Tran
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Weijun Shen
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Danling Wang
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Peter G Schultz
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - Feng Wang
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
50
|
Wang Y, Du J, Zou H, Liu Y, Zhang Y, Gonzalez J, Chao E, Lu L, Yang P, Parker H, Nguyen-Tran V, Shen W, Wang D, Schultz PG, Wang F. Multifunctional Antibody Agonists Targeting Glucagon-like Peptide-1, Glucagon, and Glucose-Dependent Insulinotropic Polypeptide Receptors. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201606321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ying Wang
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Jintang Du
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Huafei Zou
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Yan Liu
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Yuhan Zhang
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Jose Gonzalez
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Elizabeth Chao
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Lucy Lu
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Pengyu Yang
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Holly Parker
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Van Nguyen-Tran
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Weijun Shen
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Danling Wang
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Peter G. Schultz
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| | - Feng Wang
- California Institute for Biomedical Research (Calibr); 11119 N. Torrey Pines Road La Jolla CA 92037 USA
| |
Collapse
|