1
|
Subramanian D, Chin A, Shi Y, Liu GW, Langer R, Traverso G. Identification and Validation of Cyclic Peptides with Mucin-Selective, Location-Specific Binding in the Gastrointestinal Tract. ACS NANO 2025; 19:14693-14706. [PMID: 40216380 PMCID: PMC12020424 DOI: 10.1021/acsnano.4c13520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/23/2025]
Abstract
Oral drug delivery is a widely preferred method of drug administration due to its ease of use and convenience for patients. Localization of drug release in the gastrointestinal (GI) tract is important to treat localized diseases and maximize drug absorption. However, achieving drug localization in the dynamic GI tract is challenging. To address this challenge, we leveraged the geographic diversity of the GI tract by targeting its mucus layers, which coat the epithelial surfaces. These layers, composed of mucin glycoproteins, are synthesized with unique chemical compositions and expressed in different regions, making them ideal targets for drug localization. In this article, we identify cyclic peptides that bind selectively to MUC2 (in the intestines) and MUC5AC (in the stomach), serving as targeting ligands to these regions of the GI tract. We demonstrate the effectiveness of these peptides through in vitro, ex vivo, and in vivo experiments, showing that incorporating these targeting ligands can increase binding and selectivity 2-fold to the desired regions, thus potentially overcoming challenges with localizing drug distribution in oral delivery. These results indicate that cyclic peptides can be used to localize drug cargoes at certain sites in the body compared to free drugs.
Collapse
Affiliation(s)
- Deepak
A. Subramanian
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Austin Chin
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Yunhua Shi
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Gary W. Liu
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Robert Langer
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Giovanni Traverso
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division
of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
2
|
Elmaci DN, Hopping G, Hoffmann W, Muttenthaler M, Stein M. The structural integrity of human TFF1 under reducing conditions. Redox Biol 2025; 81:103534. [PMID: 39978303 PMCID: PMC11889601 DOI: 10.1016/j.redox.2025.103534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
The trefoil factor family (TFF) comprises three secretory peptides (TFF1, TFF2, TFF3) that regulate diverse physiological processes to maintain gastrointestinal mucosal integrity and homeostasis. The TFF domain is stabilized by six conserved cysteine residues forming three intramolecular disulfide bonds. In this work, we investigated human TFF1 domain stability against increasing concentrations of the reducing agent tris(2-carboxyethyl)phosphine (TCEP). Experiments revealed high resistance of the disulfide bonds within the TFF1 domain to reduction compared to two reference peptides with similar three-disulfide frameworks, namely the bovine pancreatic trypsin inhibitor (BPTI) and the peptide drug linaclotide. Full reduction of TFF1 was only achieved with a large excess of TCEP (150-fold), and no partially reduced intermediates were observed, supporting a compact TFF1 domain. This TFF1 domain stability was supported by extensive all-atom molecular dynamics simulations for a total of 24 μs of all possible combinatorial states of disulfide bond reduction. Despite minor structural and conformational changes observed upon reduction, the domain substantially retained its overall compactness and solvent exposure when only one or two disulfide bonds were removed. The reduced cysteine residues did not undergo large structural rearrangements and remained buried. The loss of covalent disulfide bonds upon reduction was counterbalanced through persistent non-covalent interactions. These molecular simulations explained why TFF1 could not be partially reduced and alkylated during the experiments despite titrating different TCEP concentrations in the presence of alkylating agents. Our findings provide the first insights into the remarkable stability of the human TFF domain under reducing conditions, supporting its functional resilience upon expression and secretion throughout the human body.
Collapse
Affiliation(s)
- Dilsah Nur Elmaci
- Molecular Simulations and Design Group, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany
| | - Gene Hopping
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Werner Hoffmann
- Institute for Molecular Biology and Medicinal Chemistry, Medical Faculty Otto von Guericke University, 39120 Magdeburg, Germany
| | - Markus Muttenthaler
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia; Institute of Biological Chemistry, University of Vienna, 1090 Vienna, Austria.
| | - Matthias Stein
- Molecular Simulations and Design Group, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany.
| |
Collapse
|
3
|
Qin L, Zhai Q, Cui Z, Li H, Guan J, Zhang Z, Xu E, Zhang X, Mao S. Elucidating structure of endogenous phospholipids on in vivo absorption of octreotide following lung administration. Eur J Pharm Biopharm 2025:114706. [PMID: 40174681 DOI: 10.1016/j.ejpb.2025.114706] [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: 12/12/2024] [Revised: 03/15/2025] [Accepted: 03/29/2025] [Indexed: 04/04/2025]
Abstract
Phospholipids as endogenous pulmonary components have received extensive attention on promoting the transmembrane absorption of peptides and proteins. However, considering their diversified structure, influence of phospholipid structural characteristics on drug absorption across lung epithelial cells, together with the underlying absorption-promoting mechanisms, remain unclear. Therefore, in this study, taking octreotide as a model drug, phospholipids with different "tail" and "head" structures were adopted in the form of blank liposomes to investigate their structural characteristics on drug absorption utilizing both 3D Transwell cell models and Sprague Dawley rats. It was demonstrated that indeed the absorption-enhancing capacity of phospholipids was their structure-dependent. Among the tail (non-polar) structures, a moderately increased alkyl chain length could facilitate drug absorption across the pulmonary epithelium, with the highest enhancement ratio observed for Dipalmitoyl Phosphatidylcholine (DPPC) containing a palmitoyl group of 16 carbons, and its apparent permeability coefficient (Papp) increased 2.4 times compared to octreotide solution. Among the head (polar) structures, charged functional groups could contribute to better drug permeation, and Dipalmitoyl Phosphatidylserine (DPPS) containing a protonated amino (NH3+) and a deprotonated carboxyl (COO-) exhibited a 4.6-fold increase in Papp compared to octreotide solution. Mechanism studies disclosed a paracellular pathway-mediated drug transport across lung epithelial cells. In summary, phospholipids can serve as biosafe absorption enhancers for pulmonary drug delivery, with the extent depending on their structure, which could provide a theoretical basis for pulmonary delivery of macromolecules for systemic absorption.
Collapse
Affiliation(s)
- Lu Qin
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qiyao Zhai
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhixiang Cui
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hongfang Li
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jian Guan
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Ziwei Zhang
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Enyu Xu
- Department of Forensic Toxicological Analysis, School of Forensic Medicine, China Medical University, Shenyang 110122, China
| | - Xin Zhang
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| |
Collapse
|
4
|
Baral KC, Choi KY. Barriers and Strategies for Oral Peptide and Protein Therapeutics Delivery: Update on Clinical Advances. Pharmaceutics 2025; 17:397. [PMID: 40284395 PMCID: PMC12030352 DOI: 10.3390/pharmaceutics17040397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 04/29/2025] Open
Abstract
Peptide and protein (PP) therapeutics are highly specific and potent biomolecules that treat chronic and complex diseases. However, their oral delivery is significantly hindered by enzymatic degradation, instability, and poor permeability through the gastrointestinal (GI) epithelium, resulting in low bioavailability. Various strategies have emerged as transformative solutions to address existing challenges, offering enhanced protection, stabilization, and absorption of PPs. These strategies primarily focus on two major challenges: protecting the PP against harsh conditions and enhancing permeation across the intestinal membrane. Innovative approaches such as pH modulation and incorporation of enzyme inhibitors are usually used to mitigate proteolytic degradation of PP during transit across the GI tract. In a similar vein, absorption enhancers and prodrug strategies facilitate epithelial transport, while targeted delivery systems focus on specific areas of the GI tract to enhance absorption. Likewise, mucus-penetrating and mucoadhesive strategies have enhanced retention and interaction with epithelial cells, effectively overcoming barriers like the mucus layer and tight epithelial junctions. Furthermore, structural modifications such as lipidation, peptide cyclization, and polyethylene glycosylation are promising alternatives to render stability, prolong circulation time, and membrane permeability. In particular, functional biomaterials, active targeting, and lymphatic transport strategies have provided new platforms for oral PP delivery. Advancing in materials science, nanotechnology, and the disruption of medical devices holds new frontiers to overcome barriers. Despite substantial advancements, the limited success in clinical translation underscores the urgency of innovative strategies. This review presents oral PPs as a promising platform, highlighting the key barriers and strategies to transform their therapeutic landscapes.
Collapse
Affiliation(s)
- Kshitis Chandra Baral
- Department of Marine Bio-Food Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea;
| | - Ki Young Choi
- Department of Marine Bio-Food Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea;
- NVience Inc., Seoul 04323, Republic of Korea
| |
Collapse
|
5
|
Gelli HP, Hedin KA, Laursen MF, Uribe RV, Sommer MOA. Enhancing intestinal absorption of a macromolecule through engineered probiotic yeast in the murine gastrointestinal tract. Trends Biotechnol 2025; 43:715-731. [PMID: 39658447 DOI: 10.1016/j.tibtech.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/20/2024] [Accepted: 10/31/2024] [Indexed: 12/12/2024]
Abstract
Oral administration of therapeutic peptides is limited by poor intestinal absorption. Use of engineered microorganisms as drug delivery vehicles can overcome the challenges faced by conventional delivery methods. The potential of engineered microorganisms to act synergistically with the therapeutics they deliver opens new horizons for noninvasive treatment modalities. This study engineered a probiotic yeast, Saccharomyces boulardii, to produce cell-penetrating peptides (CPPs) in situ for enhanced intestinal permeability. Four CPPs were integrated into the yeast chromosome: RRL helix, Shuffle, Penetramax, and PN159. In vitro tests on a Caco-2 cell model showed that three CPP-producing strains increased permeability without causing permanent damage. In vivo experiments on mice revealed that Sb PN159 administration over 10 days significantly increased FITC-dextran translocation into the bloodstream without causing inflammation. This study demonstrates, for the first time, the ability of an engineered microorganism to modulate host permeability for improved intestinal absorption of a macromolecule.
Collapse
Affiliation(s)
- Hitesh P Gelli
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Karl Alex Hedin
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Martin F Laursen
- National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ruben-Vazquez Uribe
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark; Center for Microbiology, VIB, Leuven, Belgium
| | | |
Collapse
|
6
|
Jiao Q, Huang Y, He J, Xu Y. Advances in Oral Biomacromolecule Therapies for Metabolic Diseases. Pharmaceutics 2025; 17:238. [PMID: 40006605 PMCID: PMC11859201 DOI: 10.3390/pharmaceutics17020238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic diseases like obesity and diabetes are on the rise, and therapies with biomacromolecules (such as proteins, peptides, antibodies, and oligonucleotides) play a crucial role in their treatment. However, these drugs are traditionally injected. For patients with chronic diseases (e.g., metabolic diseases), long-term injections are accompanied by inconvenience and low compliance. Oral administration is preferred, but the delivery of biomacromolecules is challenging due to gastrointestinal barriers. In this article, we introduce the available biomacromolecule drugs for the treatment of metabolic diseases. The gastrointestinal barriers to oral drug delivery and strategies to overcome these barriers are also explored. We then discuss strategies for alleviating metabolic defects, including glucose metabolism, lipid metabolism, and energy metabolism, with oral biomacromolecules such as insulin, glucagon-like peptide-1 receptor agonists, proprotein convertase subtilisin/kexin type 9 inhibitors, fibroblast growth factor 21 analogues, and peptide YY analogues.
Collapse
Affiliation(s)
- Qiuxia Jiao
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Huang
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yining Xu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
Vazquez-Uribe R, Hedin KA, Licht TR, Nieuwdorp M, Sommer MOA. Advanced microbiome therapeutics as a novel modality for oral delivery of peptides to manage metabolic diseases. Trends Endocrinol Metab 2025; 36:29-41. [PMID: 38782649 DOI: 10.1016/j.tem.2024.04.021] [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: 03/13/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
The rising prevalence of metabolic diseases calls for innovative treatments. Peptide-based drugs have transformed the management of conditions such as obesity and type 2 diabetes. Yet, challenges persist in oral delivery of these peptides. This review explores the potential of 'advanced microbiome therapeutics' (AMTs), which involve engineered microbes for delivery of peptides in situ, thereby enhancing their bioavailability. Preclinical work on AMTs has shown promise in treating animal models of metabolic diseases, including obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease. Outstanding challenges toward realizing the potential of AMTs involve improving peptide expression, ensuring predictable colonization control, enhancing stability, and managing safety and biocontainment concerns. Still, AMTs have potential for revolutionizing the treatment of metabolic diseases, potentially offering dynamic and personalized novel therapeutic approaches.
Collapse
Affiliation(s)
- Ruben Vazquez-Uribe
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Karl Alex Hedin
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Tine Rask Licht
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Max Nieuwdorp
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Morten O A Sommer
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark.
| |
Collapse
|
8
|
Çelik Tekeli M, Yalçın Y, Verdi H, Aktaş Y, Çelebi N. In vitro cellular uptake and insulin secretion studies on INS-1E cells of exendin-4-loaded self-nanoemulsifying drug delivery systems. Pharm Dev Technol 2024; 29:1101-1110. [PMID: 39474799 DOI: 10.1080/10837450.2024.2423823] [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: 06/12/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/09/2024]
Abstract
Exendin-4 (ex-4) is a peptide molecule that regulates blood glucose levels without causing hypoglycemia by providing insulin secretion from beta cells in the pancreas. Self-nanoemulsifying drug delivery systems (SNEDDS) attract attention for oral administration of therapeutic peptide/proteins because they protect therapeutic peptide/proteins from the gastric environment, reduce changes due to food effects, are easy to prepare and scale-up. Ex-4 has no commercial form that can be administered orally. In this study, the cytotoxicity, cellular uptake, and insulin secretion of ex-4 and ex-4/chymostatin (chym) SNEDDS were investigated on INS-1E rat pancreatic beta cells. The effect of ex-4 and ex-4/chym SNEDDS on cell viability in INS-1E cells increased when the dilution ratio higher. Ex-4 and ex-4/chym SNEDDS increased insulin levels in 2.8 mM (low-dose) glucose-induced INS-1E cells 2.21-fold and 2.17-fold compared to control, respectively. Ex-4 and ex-4/chym SNEDDS increased insulin levels in 16.7 mM (high dose) glucose-induced INS-1E cells compared to control, respectively. In cellular uptake studies, coumarin-6 solution penetrated the apical membrane of INS-1E cells and remained in the cytoplasm, while coumarin-6 loaded SNEDDS were visualized in the nuclei of the cell. These findings will likely be useful in the development of new formulations for the oral administration of peptides/proteins.
Collapse
Affiliation(s)
- Merve Çelik Tekeli
- Department of Pharmaceutical Technology, Erciyes University Faculty of Pharmacy, Kayseri, Turkey
| | - Yaprak Yalçın
- Department of Medical Biology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Hasibe Verdi
- Department of Medical Biology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Yeşim Aktaş
- Department of Pharmaceutical Technology, Erciyes University Faculty of Pharmacy, Kayseri, Turkey
| | - Nevin Çelebi
- Department of Pharmaceutical Technology, Baskent University Faculty of Pharmacy, Ankara, Turkey
| |
Collapse
|
9
|
Fawaz M, Sun C, Feng Y, Qirjollari A, Josien H, DeBord D, Simone A, Williamson DL, Pearson K, Gonzalez RJ, Vasicek L, Cancilla MT, Wang W, Spellman DS, Kedia K. Leveraging High-Resolution Ion Mobility-Mass Spectrometry for Cyclic Peptide Soft Spot Identification. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:2596-2607. [PMID: 38992936 DOI: 10.1021/jasms.4c00109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Cyclic peptides are an important class of molecules that gained significant attention in the field of drug discovery due to their unique pharmacological characteristics and enhanced proteolytic stability. Yet, gastrointestinal degradation remains a major hurdle in the discovery of orally bioavailable cyclic peptides. Soft spot identification (SSID) of the regions in the cyclic peptide sequence susceptible to amide hydrolysis by proteases is used in the discovery stage to guide medicinal chemistry design. SSID can be an arduous task, traditionally performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS), often resulting in complex and time-consuming manual analysis, particularly when isomeric linear peptide metabolites chromatographically coelute. Here, we present an alternative orthogonal approach that entails a high-resolution ion mobility (HRIM) system based on Structures for Lossless Ion Manipulation (SLIM) technology interfaced with quadrupole time-of-flight (QTOF) mass spectrometry to address some of the challenges associated with SSID. Two strategies were used to resolve linear isomeric peptide metabolites: labeled and label-free, both utilizing the HRIM platform. The label-free strategy leverages negative polarity to ionize the isomers which achieves better separation of the gas phase ions in the ion mobility (IM) dimension as compared to positive polarity, which is a more conventional approach when studying proteins and peptides. The second approach uses an isotope-labeled dimethyl tag on the terminal amine group, acting as a "shift reagent" to influence the mobility of isomers in the positive mode. This method resulted in baseline separation for the isomers of interest and produced unique product ions in the fragmentation spectra for unambiguous soft spot identification. Both label-free and labeled strategies demonstrated the ability to solve the challenges associated with SSID for cyclic peptides.
Collapse
Affiliation(s)
- Maria Fawaz
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Congliang Sun
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Yu Feng
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Hubert Josien
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Daniel DeBord
- MOBILion Systems, Inc., Chadds Ford, Pennsylvania 19317, United States
| | - Ashli Simone
- MOBILion Systems, Inc., Chadds Ford, Pennsylvania 19317, United States
| | | | - Kara Pearson
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Lisa Vasicek
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Mark T Cancilla
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Weixun Wang
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Komal Kedia
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
10
|
Kwak SB, Kim SJ, Kang YJ, Lee WW, Huh J, Park JW. Development of a rectally administrable Dnase1 to treat septic shock by targeting NETs. Life Sci 2024; 342:122526. [PMID: 38417543 DOI: 10.1016/j.lfs.2024.122526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/11/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
AIMS Neutrophil extracellular trap (NET), which is formed by DNA threads, induces septic shock by aggravating systemic inflammation. An intravenous administration of deoxyribonuclease is regarded as a compelling modality for treating septic shock. However, alternative routes should be chosen when cutaneous veins are all collapsed due to hypotension. In this study, we genetically engineered this enzyme to develop a rectal suppository formulation to treat septic shock. MAIN METHODS Dnase1 was mutated at two amino acid residues to increase its stability in the blood and fused with a cell-penetrating peptide CR8 to increase its absorption through the rectal mucosa, which is designated AR-CR8. The life-saving effect of AR-CR8 was evaluated in a LPS-induced shock mouse model. KEY FINDINGS AR-CR8 was shown to remove NETs effectively in human neutrophils. When AR-CR8 was administered to the mouse rectum, the deoxyribonuclease activity in the mouse serum was significantly increased. In the LPS-induced shock model, 90 % of the control mice died over 72 h after LPS injection. In contrast, the rectal administration of AR-CR8 showed a mortality rate of 30 % by 72 h after LPS injection. The Log-rank test revealed that the survival rate is significantly higher in the AR-CR8 group. The NET markers in the mouse serum were enhanced by LPS, and significantly downregulated in the AR-CR8 group. These results suggest that AR-CR8 ameliorates LPS-induced shock by degrading NETs. SIGNIFICANCE The engineered DNASE1 could be developed as a rectal suppository formulation to treat septic shock urgently at out-of-hospital places where no syringe is available.
Collapse
Affiliation(s)
- Su-Bin Kwak
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro 103, Jongno-gu, Seoul 03080, Republic of Korea
| | - Sang-Jin Kim
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro 103, Jongno-gu, Seoul 03080, Republic of Korea
| | - Yeon Jun Kang
- Laboratory of Autoimmunity and Inflammation, Department of Biomedical Sciences, Seoul National University College of Medicine, Daehak-ro 103, Jongno-gu, Seoul 03080, Republic of Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Daehak-ro 103, Jongno-gu, Seoul 03080, Republic of Korea
| | - Won-Woo Lee
- Laboratory of Autoimmunity and Inflammation, Department of Biomedical Sciences, Seoul National University College of Medicine, Daehak-ro 103, Jongno-gu, Seoul 03080, Republic of Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Daehak-ro 103, Jongno-gu, Seoul 03080, Republic of Korea
| | - June Huh
- Department of Chemical and Biological Engineering, Korea University, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jong-Wan Park
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro 103, Jongno-gu, Seoul 03080, Republic of Korea; Department of Biomedical Science, BK21-plus education program, Seoul National University College of Medicine, Daehak-ro 103, Jongno-gu, Seoul 03080, Republic of Korea; Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Daehak-ro 103, Jongno-gu, Seoul 03080, Republic of Korea.
| |
Collapse
|
11
|
Zhang E, Zhu H, Song B, Shi Y, Cao Z. Recent advances in oral insulin delivery technologies. J Control Release 2024; 366:221-230. [PMID: 38161033 PMCID: PMC10932876 DOI: 10.1016/j.jconrel.2023.12.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/23/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
With the rise in diabetes mellitus cases worldwide, oral delivery of insulin is preferred over subcutaneous insulin administration due to its good patient compliance and non-invasiveness, simplicity, and versatility. However, oral insulin delivery is hampered by various gastrointestinal barriers that result in low drug bioavailability and insufficient therapeutic efficiency. Numerous strategies have been developed to overcome these barriers and increase the bioavailability of oral insulin. Yet, no commercial oral insulin product is available to address all clinical hurdles because of various substantial obstacles related to the structural organization and physiological function of the gastrointestinal tract. Herein, we discussed the significant physiological barriers (including chemical, enzymatic, and physical barriers) that hinder the transportation and absorption of orally delivered insulin. Then, we showcased recent significant and innovative advances in oral insulin delivery technologies. Finally, we concluded the review with remarks on future perspectives on oral insulin delivery technologies and potential challenges for forthcoming clinical translation of oral insulin delivery technologies.
Collapse
Affiliation(s)
- Ershuai Zhang
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Hui Zhu
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Boyi Song
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Yuanjie Shi
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Zhiqiang Cao
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
12
|
To D, Kali G, Haddadzadegan S, Jörgensen AM, Nigl K, Ricci F, Bernkop-Schnürch A. Power-Up for Mucoadhesiveness: Two Generations of Thiolated Surfactants for Enhanced Sticky Nanoemulsions. ACS Biomater Sci Eng 2023; 9:6797-6804. [PMID: 37996083 PMCID: PMC10716821 DOI: 10.1021/acsbiomaterials.3c01207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023]
Abstract
Nanoemulsions can be tuned toward enhanced gastro-intestinal retention time by incorporating thiolated surfactants into their surface. Tailoring the chemical reactivity of the thiol headgroup has major influence on mucoadhesive features of the nanoemulsion. Two generations of thiolated surfactants were synthetically derived from PEG-40-stearate featuring either a free thiol group or an S-protected thiol group. The surfactants were characterized regarding critical micelle concentration (CMC), hemolytic activity, and cytotoxicity. Subsequently, they were incorporated into nanoemulsions and the resulting nanoemulsions were characterized regarding particle size, polydispersity index (PDI), zeta potential, and time-dependent stability. Afterward, mucosal interactions as well as mucoadhesion on porcine intestinal mucosa were investigated. Successful synthesis of Cysteine-PEG-40-stearate (CYS-PEG-40-stearate) and MNA-Cysteine-PEG-40-stearate (MNA-CYS-PEG-40-stearate) was confirmed by 1H NMR spectroscopy. Both chemical modifications led to slightly elevated CMC values while preserving low cytotoxicity and hemotoxicity. Incorporation into nanoemulsions had minor influence on overall physical particle characteristics, while interactions with mucus and mucoadhesiveness of the nanoemulsions were drastically improved resulting in the rank order PEG-40-stearate < CYS-PEG-40-stearate < MNA-CYS-PEG-40-stearate. Accordingly, thiolated surfactants, especially S-protected derivatives, are versatile tools to generate highly mucoadhesive nanoemulsions.
Collapse
Affiliation(s)
- Dennis To
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Gergely Kali
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Soheil Haddadzadegan
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Arne Matteo Jörgensen
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Katharina Nigl
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Fabrizio Ricci
- Thiomatrix
Forschungs- und Beratungs GmbH, Trientlgasse 65, 6020 Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| |
Collapse
|
13
|
Lyons B, Balkaran JPR, Dunn-Lawless D, Lucian V, Keller SB, O’Reilly CS, Hu L, Rubasingham J, Nair M, Carlisle R, Stride E, Gray M, Coussios C. Sonosensitive Cavitation Nuclei-A Customisable Platform Technology for Enhanced Therapeutic Delivery. Molecules 2023; 28:7733. [PMID: 38067464 PMCID: PMC10708135 DOI: 10.3390/molecules28237733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Ultrasound-mediated cavitation shows great promise for improving targeted drug delivery across a range of clinical applications. Cavitation nuclei-sound-sensitive constructs that enhance cavitation activity at lower pressures-have become a powerful adjuvant to ultrasound-based treatments, and more recently emerged as a drug delivery vehicle in their own right. The unique combination of physical, biological, and chemical effects that occur around these structures, as well as their varied compositions and morphologies, make cavitation nuclei an attractive platform for creating delivery systems tuned to particular therapeutics. In this review, we describe the structure and function of cavitation nuclei, approaches to their functionalization and customization, various clinical applications, progress toward real-world translation, and future directions for the field.
Collapse
Affiliation(s)
- Brian Lyons
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Joel P. R. Balkaran
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Darcy Dunn-Lawless
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Veronica Lucian
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Sara B. Keller
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Colm S. O’Reilly
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX1 3PJ, UK;
| | - Luna Hu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Jeffrey Rubasingham
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Malavika Nair
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| |
Collapse
|
14
|
Zhang S, Zhu C, Huang W, Liu H, Yang M, Zeng X, Zhang Z, Liu J, Shi J, Hu Y, Shi X, Wang ZH. Recent progress of micro/nanomotors to overcome physiological barriers in the gastrointestinal tract. J Control Release 2023; 360:514-527. [PMID: 37429360 DOI: 10.1016/j.jconrel.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/12/2023]
Abstract
Oral administration is a convenient administration route for gastrointestinal disease therapy with good patient compliance. But the nonspecific distribution of the oral drugs may cause serious side effects. In recent years, oral drug delivery systems (ODDS) have been applied to deliver the drugs to the gastrointestinal disease sites with decreased side effects. However, the delivery efficiency of ODDS is tremendously limited by physiological barriers in the gastrointestinal sites, such as the long and complex gastrointestinal tract, mucus layer, and epithelial barrier. Micro/nanomotors (MNMs) are micro/nanoscale devices that transfer various energy sources into autonomous motion. The outstanding motion characteristics of MNMs inspired the development of targeted drug delivery, especially the oral drug delivery. However, a comprehensive review of oral MNMs for the gastrointestinal diseases therapy is still lacking. Herein, the physiological barriers of ODDS were comprehensively reviewed. Afterward, the applications of MNMs in ODDS for overcoming the physiological barriers in the past 5 years were highlighted. Finally, future perspectives and challenges of MNMs in ODDS are discussed as well. This review will provide inspiration and direction of MNMs for the therapy of gastrointestinal diseases, pushing forward the clinical application of MNMs in oral drug delivery.
Collapse
Affiliation(s)
- Shuhao Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Chaoran Zhu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Wanting Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Hua Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Mingzhu Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Xuejiao Zeng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Yurong Hu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China.
| | - Xiufang Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China.
| | - Zhi-Hao Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China.
| |
Collapse
|
15
|
Bajoria S, Antunez LR, Kumru OS, Klempner M, Wang Y, Cavacini LA, Joshi SB, Volkin DB. Formulation Studies to Develop Low-Cost, Orally-Delivered Secretory IgA Monoclonal Antibodies for Passive Immunization Against Enterotoxigenic Escherichia coli. J Pharm Sci 2023; 112:1832-1844. [PMID: 37040833 DOI: 10.1016/j.xphs.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/06/2023] [Accepted: 04/06/2023] [Indexed: 04/13/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a common cause for diarrheal infections in children in low- and middle-income countries (LMICs). To date, no ETEC vaccine candidates have been approved. Passive immunization with low-cost, oral formulations of secretory IgA (sIgA) against ETEC is an alternative approach to protect high-risk populations in LMICs. Using a model sIgA monoclonal antibody (anti-LT sIgA2-mAb), the stability profiles of different formulations were assessed during storage and in in vitro digestion models (mimicking in vivo oral delivery). First, by employing various physicochemical techniques and a LT-antigen binding assay, three formulations with varying acid-neutralizing capacity (ANC) were evaluated to stabilize sIgA2-mAb during stress studies (freeze-thaw, agitation, elevated temperature) and during exposure to gastric phase digestion. Next, a low-volume, in vitro intestinal digestion model was developed to screen various additives to stabilize sIgA2-mAb in the intestinal phase. Finally, combinations of high ANC buffers and decoy proteins were assessed to collectively protect sIgA2-mAb during in vitro sequential (stomach to intestine) digestion. Based on the results, we demonstrate the feasibility of low-cost, 'single-vial', liquid formulations of sIgA-mAbs delivered orally after infant feeding for passive immunization, and we suggest future work based on a combination of in vitro and in vivo stability considerations.
Collapse
Affiliation(s)
- Sakshi Bajoria
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center (VAFC), University of Kansas, Lawrence, KS 66047, USA
| | - Lorena R Antunez
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center (VAFC), University of Kansas, Lawrence, KS 66047, USA
| | - Ozan S Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center (VAFC), University of Kansas, Lawrence, KS 66047, USA
| | - Mark Klempner
- MassBiologics of the University of Massachusetts Chan Medical School, Boston, MA 02126, USA
| | - Yang Wang
- MassBiologics of the University of Massachusetts Chan Medical School, Boston, MA 02126, USA
| | - Lisa A Cavacini
- MassBiologics of the University of Massachusetts Chan Medical School, Boston, MA 02126, USA
| | - Sangeeta B Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center (VAFC), University of Kansas, Lawrence, KS 66047, USA
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center (VAFC), University of Kansas, Lawrence, KS 66047, USA.
| |
Collapse
|
16
|
Masloh S, Culot M, Gosselet F, Chevrel A, Scapozza L, Zeisser Labouebe M. Challenges and Opportunities in the Oral Delivery of Recombinant Biologics. Pharmaceutics 2023; 15:pharmaceutics15051415. [PMID: 37242657 DOI: 10.3390/pharmaceutics15051415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Recombinant biological molecules are at the cutting-edge of biomedical research thanks to the significant progress made in biotechnology and a better understanding of subcellular processes implicated in several diseases. Given their ability to induce a potent response, these molecules are becoming the drugs of choice for multiple pathologies. However, unlike conventional drugs which are mostly ingested, the majority of biologics are currently administered parenterally. Therefore, to improve their limited bioavailability when delivered orally, the scientific community has devoted tremendous efforts to develop accurate cell- and tissue-based models that allow for the determination of their capacity to cross the intestinal mucosa. Furthermore, several promising approaches have been imagined to enhance the intestinal permeability and stability of recombinant biological molecules. This review summarizes the main physiological barriers to the oral delivery of biologics. Several preclinical in vitro and ex vivo models currently used to assess permeability are also presented. Finally, the multiple strategies explored to address the challenges of administering biotherapeutics orally are described.
Collapse
Affiliation(s)
- Solene Masloh
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Maxime Culot
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
| | - Anne Chevrel
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Magali Zeisser Labouebe
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| |
Collapse
|
17
|
Khongkow M, Rimsueb N, Jantimaporn A, Janyaphisan T, Woraprayote W, Visessanguan W, Ruktanonchai UR. Cationic liposome of hen egg white lysozyme for enhanced its stability, activity and accessibility in gastro-intestinal tract. FOOD BIOSCI 2023. [DOI: 10.1016/j.fbio.2023.102470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
18
|
Liang B, Li R, Li L, Tang M, Li X, Su C, Liao H. Silver-promoted solid-phase guanidinylation enables the first synthesis of arginine glycosylated Samoamide A cyclopeptide analogue. Front Chem 2023; 10:1040216. [PMID: 36688048 PMCID: PMC9846560 DOI: 10.3389/fchem.2022.1040216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023] Open
Abstract
Cyclization and glycosylation serve as effective approaches for enhancing the drug properties of peptides. Distinct from typical glycosylation, atypical arginine N-glycosylation has drawn increasing attention due to its fundamental role in various cellular procedures and signaling pathways. We previously developed a robust strategy for constructing arginine N-glycosylated peptides characterized by silver-promoted solid-phase guanidinylation. Modeled after cyclic octapeptide Samoamide A, an antitumor peptide composed of eight hydrophobic amino acids extracted from cyanobacteria, herein we first performed arginine scanning to determine an optimal position for replacement with arginine. Consequently, the first synthesis of arginine glycosylated Samoamide A cyclopeptide analogue was described combining solid-phase glycosylation with solution-phase cyclization. The resultant SA-HH-TT displayed enhanced water solubility compared with the non-glycosylated SA-HH-TT. Notably, our method provides a universal strategy for synthesizing arginine N-glycosylated cyclopeptides.
Collapse
Affiliation(s)
- Bingxin Liang
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Rong Li
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Linji Li
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Ming Tang
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, Shanghai, China,*Correspondence: Xiang Li, ; Chunli Su, ; Hongli Liao,
| | - Chunli Su
- School of Public Health, Chengdu Medical College, Chengdu, China,*Correspondence: Xiang Li, ; Chunli Su, ; Hongli Liao,
| | - Hongli Liao
- School of Pharmacy, Chengdu Medical College, Chengdu, China,*Correspondence: Xiang Li, ; Chunli Su, ; Hongli Liao,
| |
Collapse
|
19
|
Enhancement of oral bioavailability of insulin using a combination of surface-modified inclusion complex and SNEDDS. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.120641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Qin L, Cui Z, Wu Y, Wang H, Zhang X, Guan J, Mao S. Challenges and Strategies to Enhance the Systemic Absorption of Inhaled Peptides and Proteins. Pharm Res 2022; 40:1037-1055. [DOI: 10.1007/s11095-022-03435-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/07/2022] [Indexed: 11/17/2022]
|
21
|
Anand U, Bandyopadhyay A, Jha NK, Pérez de la Lastra JM, Dey A. Translational aspect in peptide drug discovery and development: An emerging therapeutic candidate. Biofactors 2022; 49:251-269. [PMID: 36326181 DOI: 10.1002/biof.1913] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 08/11/2022] [Indexed: 11/06/2022]
Abstract
In the last two decades, protein-protein interactions (PPIs) have been used as the main target for drug development. However, with larger or superficial binding sites, it has been extremely difficult to disrupt PPIs with small molecules. On the other hand, intracellular PPIs cannot be targeted by antibodies that cannot penetrate the cell membrane. Peptides that have a combination of conformational rigidity and flexibility can be used to target difficult binding interfaces with appropriate binding affinity and specificity. Since the introduction of insulin nearly a century ago, more than 80 peptide drugs have been approved to treat a variety of diseases. These include deadly diseases such as cancer and human immunodeficiency virus infection. It is also useful against diabetes, chronic pain, and osteoporosis. Today, more research is being done on these drugs as lessons learned from earlier approaches, which are still valid today, complement newer approaches such as peptide display libraries. At the same time, integrated genomics and peptide display libraries are new strategies that open new avenues for peptide drug discovery. The purpose of this review is to examine the problems in elucidating the peptide-protein recognition mechanism. This is important to develop peptide-based interventions that interfere with endogenous protein interactions. New approaches are being developed to improve the binding affinity and specificity of existing approaches and to develop peptide agents as potentially useful drugs. We also highlight the key challenges that must be overcome in peptide drug development to realize their potential and provide an overview of recent trends in peptide drug development. In addition, we take an in-depth look at early efforts in human hormone discovery, smart medicinal chemistry and design, natural peptide drugs, and breakthrough advances in molecular biology and peptide chemistry.
Collapse
Affiliation(s)
- Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, Punjab, India
- Department of Biotechnology, School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - José M Pérez de la Lastra
- Biotechnology of Macromolecules Research Group, Instituto de Productos Naturales y Agrobiología, IPNA-CSIC, Tenerife, Spain
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| |
Collapse
|
22
|
He S, Yang Z, Li X, Wu H, Zhang L, Wang J, Shan A. Optimized proteolytic resistance motif (DabW)-based U1-2WD: A membrane-induced self-aggregating peptide to trigger bacterial agglutination and death. Acta Biomater 2022; 153:540-556. [PMID: 36162762 DOI: 10.1016/j.actbio.2022.09.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 12/13/2022]
Abstract
The biggest application bottleneck of antimicrobial peptides (AMPs) is the low oral bioavailability caused by the poor stability of digestive enzymes in the gastrointestinal tract. However, the research methods and evaluation criteria of available studies about anti-proteolytic strategies are not uniform and far from the actual environment in vivo. Here, we developed a research system and evaluation criteria for proteolytic resistance and systematically evaluated the effectiveness of different strategies for improving the protease stability of AMPs on the same platform for the first time. After a comprehensive analysis, Dab modification is identified as the most effective strategy to improve the trypsin stability of AMPs. By further modulating the proteolytic resistance optimization motif (DabW)n, U1-2WD is obtained with ideal stability and antimicrobial properties in vivo and in vitro. Notably, U1-2WD has a unique antibacterial mechanism, which forms amorphous aggregates in the bacteria environment to trigger the agglutination of bacterial cells to prevent bacterial escape. It then kills bacteria by disrupting bacterial membranes and inhibiting bacterial energy metabolism. Overall, our work has led to a new understanding of the effectiveness of proteolytic resistance strategies and accelerated the development of anti-proteolytic AMPs to combat multidrug-resistant bacterial infections. STATEMENT OF SIGNIFICANCE: We developed research system and evaluation criteria for proteolytic resistance and systematically evaluated the effectiveness of different strategies for improving protease stability of AMPs on the same platform for the first time. we found effective strategies to resist trypsin hydrolysis: modification with backbone (β-Arg), D-enantiomer (D-Arg) and L-2,4-diaminobutanoic acid (Dab). Further, the proteolytic resistance optimization motif (DabW)n was designed. When n=3, derivative U1-2WD was obtained with desirable stability and antimicrobial properties in vivo and in vitro. Notably, U1-2WD has a unique antibacterial mechanism, which can self-aggregate into amorphous aggregates in the bacteria environment to mediate the agglutination and sedimentation of bacterial cells to prevent bacterial escape, and then kill bacteria by destroying bacterial membranes and inhibiting bacterial energy metabolism.
Collapse
Affiliation(s)
- Shiqi He
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Zhanyi Yang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xuefeng Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Hua Wu
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Licong Zhang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Jiajun Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China.
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China.
| |
Collapse
|
23
|
Modification and preparation of four natural hydrogels and their application in biopharmaceutical delivery. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04412-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
24
|
Abstract
Peptides have traditionally been perceived as poor drug candidates due to unfavorable characteristics mainly regarding their pharmacokinetic behavior, including plasma stability, membrane permeability and circulation half-life. Nonetheless, in recent years, general strategies to tackle those shortcomings have been established, and peptides are subsequently gaining increasing interest as drugs due to their unique ability to combine the advantages of antibodies and small molecules. Macrocyclic peptides are a special focus of drug development efforts due to their ability to address so called ‘undruggable’ targets characterized by large and flat protein surfaces lacking binding pockets. Here, the main strategies developed to date for adapting peptides for clinical use are summarized, which may soon help usher in an age highly shaped by peptide-based therapeutics. Nonetheless, limited membrane permeability is still to overcome before peptide therapeutics will be broadly accepted.
Collapse
|
25
|
Eng H, Dantonio AL, Kadar EP, Obach RS, Di L, Lin J, Patel NC, Boras B, Walker GS, Novak JJ, Kimoto E, Singh RSP, Kalgutkar AS. Disposition of Nirmatrelvir, an Orally Bioavailable Inhibitor of SARS-CoV-2 3C-Like Protease, across Animals and Humans. Drug Metab Dispos 2022; 50:576-590. [PMID: 35153195 DOI: 10.1124/dmd.121.000801] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/01/2022] [Indexed: 02/13/2025] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) 3C-like protease inhibitor PF-07321332 (nirmatrelvir), in combination with ritonavir (Paxlovid), was recently granted emergency use authorization by multiple regulatory agencies for the treatment of coronavirus disease 2019 (COVID-19) in adults and pediatric patients. Disposition studies on nirmatrelvir in animals and in human reagents, which were used to support clinical studies, are described herein. Plasma clearance was moderate in rats (27.2 ml/min per kg) and monkeys (17.1 ml/min per kg), resulting in half-lives of 5.1 and 0.8 hours, respectively. The corresponding oral bioavailability was moderate in rats (34%-50%) and low in monkeys (8.5%), primarily due to oxidative metabolism along the gastrointestinal tract in this species. Nirmatrelvir demonstrated moderate plasma protein binding in rats, monkeys, and humans with mean unbound fractions ranging from 0.310 to 0.478. The metabolism of nirmatrelvir was qualitatively similar in liver microsomes and hepatocytes from rats, monkeys, and humans; prominent metabolites arose via cytochrome P450 (CYP450)-mediated oxidations on the P1 pyrrolidinone ring, P2 6,6-dimethyl-3-azabicyclo[3.1.0]hexane, and the tertiary-butyl group at the P3 position. Reaction phenotyping studies in human liver microsomes revealed that CYP3A4 was primarily responsible (fraction metabolized = 0.99) for the oxidative metabolism of nirmatrelvir. Minor clearance mechanisms involving renal and biliary excretion of unchanged nirmatrelvir were also noted in animals and in sandwich-cultured human hepatocytes. Nirmatrelvir was a reversible and time-dependent inhibitor as well as inducer of CYP3A activity in vitro. First-in-human pharmacokinetic studies have demonstrated a considerable boost in the oral systemic exposure of nirmatrelvir upon coadministration with the CYP3A4 inhibitor ritonavir, consistent with the predominant role of CYP3A4 in nirmatrelvir metabolism. SIGNIFICANCE STATEMENT: The manuscript describes the preclinical disposition, metabolism, and drug-drug interaction potential of PF-07321332 (nirmatrelvir), an orally active peptidomimetic-based inhibitor of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) 3CL protease, which has been granted emergency use authorization by multiple regulatory agencies around the globe for the treatment of coronavirus disease 2019 (COVID-19) in COVID-19-positive adults and pediatric patients who are at high risk for progression to severe COVID-19, including hospitalization or death.
Collapse
Affiliation(s)
- Heather Eng
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Alyssa L Dantonio
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Eugene P Kadar
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - R Scott Obach
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Li Di
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Jian Lin
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Nandini C Patel
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Britton Boras
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Gregory S Walker
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Jonathan J Novak
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Emi Kimoto
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Ravi Shankar P Singh
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| | - Amit S Kalgutkar
- Pfizer Worldwide Research, Development, and Medical, Groton, Connecticut (H.E., A.L.D., E.P.K., R.S.O., L.D., J.L., G.S.W., J.J.N., E.K.); Pfizer Worldwide Research and Development, Cambridge, Massachusetts (N.C.P., R.S.P.S., A.S.K.); and Pfizer Worldwide Research and Development, La Jolla, California (B.B.)
| |
Collapse
|
26
|
Wang M, Wang C, Ren S, Pan J, Wang Y, Shen Y, Zeng Z, Cui H, Zhao X. Versatile Oral Insulin Delivery Nanosystems: From Materials to Nanostructures. Int J Mol Sci 2022; 23:3362. [PMID: 35328783 PMCID: PMC8952690 DOI: 10.3390/ijms23063362] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/24/2022] [Accepted: 02/27/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetes is a chronic metabolic disease characterized by lack of insulin in the body leading to failure of blood glucose regulation. Diabetes patients usually need frequent insulin injections to maintain normal blood glucose levels, which is a painful administration manner. Long-term drug injection brings great physical and psychological burden to diabetic patients. In order to improve the adaptability of patients to use insulin and reduce the pain caused by injection, the development of oral insulin formulations is currently a hot and difficult topic in the field of medicine and pharmacy. Thus, oral insulin delivery is a promising and convenient administration method to relieve the patients. However, insulin as a peptide drug is prone to be degraded by digestive enzymes. In addition, insulin has strong hydrophilicity and large molecular weight and extremely low oral bioavailability. To solve these problems in clinical practice, the oral insulin delivery nanosystems were designed and constructed by rational combination of various nanomaterials and nanotechnology. Such oral nanosystems have the advantages of strong adaptability, small size, convenient processing, long-lasting pharmaceutical activity, and drug controlled-release, so it can effectively improve the oral bioavailability and efficacy of insulin. This review summarizes the basic principles and recent progress in oral delivery nanosystems for insulin, including physiological absorption barrier of oral insulin and the development of materials to nanostructures for oral insulin delivery nanosystems.
Collapse
Affiliation(s)
| | | | | | | | | | - Yue Shen
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (M.W.); (C.W.); (S.R.); (J.P.); (Y.W.); (Z.Z.); (H.C.)
| | | | | | - Xiang Zhao
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (M.W.); (C.W.); (S.R.); (J.P.); (Y.W.); (Z.Z.); (H.C.)
| |
Collapse
|
27
|
Fang YQ, Chen T, Huang G, Ni S, Dang L. Reaction mechanism for copper catalyzed functionalization of unsaturated side chains of amides via domino rearrangement. J Organomet Chem 2022. [DOI: 10.1016/j.jorganchem.2021.122233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
28
|
Oral delivery of therapeutic peptides and proteins: Technology landscape of lipid-based nanocarriers. Adv Drug Deliv Rev 2022; 182:114097. [PMID: 34999121 DOI: 10.1016/j.addr.2021.114097] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/04/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022]
Abstract
The oral administration of therapeutic peptides and proteins is favoured from a patient and commercial point of view. In order to reach the systemic circulation after oral administration, these drugs have to overcome numerous barriers including the enzymatic, sulfhydryl, mucus and epithelial barrier. The development of oral formulations for therapeutic peptides and proteins is therefore necessary. Among the most promising formulation approaches are lipid-based nanocarriers such as oil-in-water nanoemulsions, self-emulsifying drug delivery systems (SEDDS), solid lipid nanoparticles (SLN), nanostructured lipid carriers (NLC), liposomes and micelles. As the lipophilic character of therapeutic peptides and proteins can be tremendously increased such as by the formation of hydrophobic ion pairs (HIP) with hydrophobic counter ions, they can be incorporated in the lipophilic phase of these carriers. Since gastrointestinal (GI) peptidases as well as sulfhydryl compounds such as glutathione and dietary proteins are too hydrophilic to enter the lipophilic phase of these carriers, the incorporated therapeutic peptide or protein is protected towards enzymatic degradation as well as unintended thiol/disulfide exchange reactions. Stability of lipid-based nanocarriers towards lipases can be provided by the use to excipients that are not or just poorly degraded by these enzymes. Nanocarriers with a size <200 nm and a mucoinert surface such as PEG or zwitterionic surfaces exhibit high mucus permeating properties. Having reached the underlying absorption membrane, lipid-based nanocarriers enable paracellular and lymphatic drug uptake, induce endocytosis and transcytosis or simply fuse with the cell membrane releasing their payload into the systemic circulation. Numerous in vivo studies provide evidence for the potential of these delivery systems. Within this review we provide an overview about the different barriers for oral peptide and protein delivery, highlight the progress made on lipid-based nanocarriers in order to overcome them and discuss strengths and weaknesses of these delivery systems in comparison to other technologies.
Collapse
|
29
|
Burshtein G, Itin C, Tang JCY, Galitzer H, Fraser WD, Schwartz P. The combined effect of permeation enhancement and proteolysis inhibition on the systemic exposure of orally administrated peptides: Salcaprozate sodium, soybean trypsin inhibitor, and teriparatide study in pigs. INTERNATIONAL JOURNAL OF PHARMACEUTICS-X 2021; 3:100097. [PMID: 34704013 PMCID: PMC8524144 DOI: 10.1016/j.ijpx.2021.100097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/21/2022]
Abstract
Oral delivery of peptides and proteins is hindered by their rapid proteolysis in the gastrointestinal tract and their inability to permeate biological membranes. Various drug delivery approaches are being investigated and implemented to overcome these obstacles. In the discussed study conducted in pigs, an investigation was undertaken to assess the effect of combination of a permeation enhancer – salcaprozate sodium, and a proteolysis inhibitor – soybean trypsin inhibitor, on the systemic exposure of the peptide teriparatide, following intraduodenal administration. Results demonstrate that this combination achieves significantly higher Cmax and AUC (~10- and ~20-fold respectively) compared to each of these methodologies on their own. It was thus concluded that an appropriate combination of different technological approaches may considerably contribute to an efficient oral delivery of biological macromolecules. Soybean trypsin inhibitor (SBTI) protects hPTH(1–34) from proteolysis in the intestine. SNAC/SBTI combination significantly raises plasma exposure of oral hPTH(1–34). Oral formulation hPTH(1–34)/SNAC/SBTI befits the PK profile for osteoporosis treatment. Endoscopic intraduodenal delivery in pigs enables investigation of absorption mechanisms.
Collapse
Affiliation(s)
| | - Constantin Itin
- Entera Bio Ltd., Jerusalem BioPark, Jerusalem 9112002, Israel
| | - Jonathan C Y Tang
- Bioanalytical Facility, Biomedical Research Centre, Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Hillel Galitzer
- Entera Bio Ltd., Jerusalem BioPark, Jerusalem 9112002, Israel
| | - William D Fraser
- Bioanalytical Facility, Biomedical Research Centre, Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK.,Departments of Endocrinology and Clinical Biochemistry, Norfolk and Norwich University Hospital, Norwich, UK
| | | |
Collapse
|
30
|
Katimba HA, Wang R, Cheng C. Current findings support the potential use of bioactive peptides in enhancing zinc absorption in humans. Crit Rev Food Sci Nutr 2021:1-21. [PMID: 34708681 DOI: 10.1080/10408398.2021.1996328] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
More than two billion people around the world are affected by zinc deficiency, mainly due to the inadequate intake and absorption of zinc. Based on recent research findings, the bioactive peptides could potentially be used to combat zinc deficiency particularly due to their Zinc chelating ability. The main aim of this review was to present current findings, supporting the potential use of bioactive peptides based on their ability to enhance zinc absorption. In-vivo, in-vitro, and ex-vivo studies have demonstrated that zinc chelating peptides can enhance the retention, transportation, and absorption of zinc. Comparative studies on zinc bioavailability from protein hydrolysates and zinc salts have demonstrated that the protein hydrolysates-zinc complexes are more bioavailable than the zinc salts. Data from the structure-function relationship of zinc chelating peptides suggest that the zinc chelating capacities of peptides increase in the following order; the position of zinc chelator > zinc chelator strength > abundance of zinc chelators > net charge > molecular weight. In addition, the transport mechanism of peptide-zinc complex is hypothesized, and the potential use of bioactive peptides based on their safety and taste and limitations to their commercialization are also discussed.
Collapse
Affiliation(s)
- Hija Athman Katimba
- Department of Food Science and Engineering, Harbin Institute of Technology, Harbin, China.,Department of Food Science and Engineering, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Rongchun Wang
- Department of Food Science and Engineering, Harbin Institute of Technology, Harbin, China
| | - Cuilin Cheng
- Department of Food Science and Engineering, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
31
|
Yang Z, He S, Wu H, Yin T, Wang L, Shan A. Nanostructured Antimicrobial Peptides: Crucial Steps of Overcoming the Bottleneck for Clinics. Front Microbiol 2021; 12:710199. [PMID: 34475862 PMCID: PMC8406695 DOI: 10.3389/fmicb.2021.710199] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
The security issue of human health is faced with dispiriting threats from multidrug-resistant bacteria infections induced by the abuse and misuse of antibiotics. Over decades, the antimicrobial peptides (AMPs) hold great promise as a viable alternative to treatment with antibiotics due to their peculiar antimicrobial mechanisms of action, broad-spectrum antimicrobial activity, lower drug residue, and ease of synthesis and modification. However, they universally express a series of disadvantages that hinder their potential application in the biomedical field (e.g., low bioavailability, poor protease resistance, and high cytotoxicity) and extremely waste the abundant resources of AMP database discovered over the decades. For all these reasons, the nanostructured antimicrobial peptides (Ns-AMPs), based on a variety of nanosystem modification, have made up for the deficiencies and pushed the development of novel AMP-based antimicrobial therapies. In this review, we provide an overview of the advantages of Ns-AMPs in improving therapeutic efficacy and biological stability, reducing side effects, and gaining the effect of organic targeting and drug controlled release. Then the different material categories of Ns-AMPs are described, including inorganic material nanosystems containing AMPs, organic material nanosystems containing AMPs, and self-assembled AMPs. Additionally, this review focuses on the Ns-AMPs for the effect of biological activities, with emphasis on antimicrobial activity, biosecurity, and biological stability. The "state-of-the-art" antimicrobial modes of Ns-AMPs, including controlled release of AMPs under a specific environment or intrinsic antimicrobial properties of Ns-AMPs, are also explicated. Finally, the perspectives and conclusions of the current research in this field are also summarized.
Collapse
Affiliation(s)
| | | | | | | | | | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
32
|
Liu L, Yao W, Xie X, Gao J, Lu X. pH-sensitive dual drug loaded janus nanoparticles by oral delivery for multimodal analgesia. J Nanobiotechnology 2021; 19:235. [PMID: 34362394 PMCID: PMC8348996 DOI: 10.1186/s12951-021-00974-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/26/2021] [Indexed: 01/15/2023] Open
Abstract
Background Based on the concept of “multimodal analgesia”, a novel dual drug delivery system was designed to achieve synergistic analgesia between najanajaatra venom protein (αCT) and resveratrol (Res). In order to meet the joint loading of two drugs with different physicochemical properties without affecting each other, an oral Janus nanoparticle (JNP) with a unique cavity structure and synergistic drug delivery was constructed using an improved double emulsion solvent evaporation method, and combined with low-molecular-weight chitosan/sodium alginate and PLGA to achieve its pH-responsive. Results The synthesized αCT/Res-JNPs are homogeneous in shape, with a two-compartment structure, approximately 230 nm in size, and zeta potential of 23.6 mV. Drug release assayed in vitro show that JNP was stable in simulated gastric juice (pH = 1.2) but was released in phosphate buffer saline (pH = 7.4). After intragastric administration in rats, PK evaluation showed that αCT/Res-JNPs could significantly improve the oral bioavailability, and the simultaneous encapsulation of the two drugs had no significant interaction on PK parameters. An obvious synergistic analgesic effects of αCT/Res-JNPs was confirmed in a spinal cord injury and acute pain model. Confocal laser scanning microscopy and single-pass intestinal perfusion model provided strong evidence that αCT/Res-JNPs could pass through intestinal epithelial cells, and the endocytosis pathway was mainly involved in the mediation and pinocytosis of reticulin. The concentrations of αCT and Res from αCT/Res-JNP in lymphatic transport were only about 8.72% and 6.08% of their blood concentrations at 1 h, respectively, which indicated that lymphatic transport in the form of JNP has limited advantages in improving the oral bioavailability of Res and αCT. Cellular uptake efficiency at 4 h was about 10–15% in Caco-2 cell lines for αCT/Res-JNP, but was reduced to 7% in Caco-2/HT29-MTX co-culture models due to the hindrance by the mucus layers. Approximately 12–17% of αCT/Res-JNP were transported across Caco-2/HT29-MTX/Raji monolayers. The cumulative absorption of JNP in three cell models was higher than that of free drug. Conclusions This study investigated the contribution of Janus nanoparticles in oral absorption, and provide a new perspective for oral administration and analgesic treatment of dual drug delivery system containing peptide drugs. Graphic Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00974-6.
Collapse
Affiliation(s)
- Lin Liu
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, PR China.,Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Wendong Yao
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University, 310018, Hangzhou, PR China
| | - Xiaowei Xie
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, PR China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Xiaoyang Lu
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, PR China.
| |
Collapse
|
33
|
Zhu Q, Chen Z, Paul PK, Lu Y, Wu W, Qi J. Oral delivery of proteins and peptides: Challenges, status quo and future perspectives. Acta Pharm Sin B 2021; 11:2416-2448. [PMID: 34522593 PMCID: PMC8424290 DOI: 10.1016/j.apsb.2021.04.001] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/29/2021] [Accepted: 02/12/2021] [Indexed: 12/24/2022] Open
Abstract
Proteins and peptides (PPs) have gradually become more attractive therapeutic molecules than small molecular drugs due to their high selectivity and efficacy, but fewer side effects. Owing to the poor stability and limited permeability through gastrointestinal (GI) tract and epithelia, the therapeutic PPs are usually administered by parenteral route. Given the big demand for oral administration in clinical use, a variety of researches focused on developing new technologies to overcome GI barriers of PPs, such as enteric coating, enzyme inhibitors, permeation enhancers, nanoparticles, as well as intestinal microdevices. Some new technologies have been developed under clinical trials and even on the market. This review summarizes the history, the physiological barriers and the overcoming approaches, current clinical and preclinical technologies, and future prospects of oral delivery of PPs.
Collapse
Key Words
- ASBT, apical sodium-dependent bile acid transporter
- BSA, bovine serum albumin
- CAGR, compound annual growth
- CD, Crohn's disease
- COPD, chronic obstructive pulmonary disease
- CPP, cell penetrating peptide
- CaP, calcium phosphate
- Clinical
- DCs, dendritic cells
- DDVAP, desmopressin acetate
- DTPA, diethylene triamine pentaacetic acid
- EDTA, ethylene diamine tetraacetic acid
- EPD, empirical phase diagrams
- EPR, electron paramagnetic resonance
- Enzyme inhibitor
- FA, folic acid
- FDA, U.S. Food and Drug Administration
- FcRn, Fc receptor
- GALT, gut-associated lymphoid tissue
- GI, gastrointestinal
- GIPET, gastrointestinal permeation enhancement technology
- GLP-1, glucagon-like peptide 1
- GRAS, generally recognized as safe
- HBsAg, hepatitis B surface antigen
- HPMCP, hydroxypropyl methylcellulose phthalate
- IBD, inflammatory bowel disease
- ILs, ionic liquids
- LBNs, lipid-based nanoparticles
- LMWP, low molecular weight protamine
- MCT-1, monocarborxylate transporter 1
- MSNs, mesoporous silica nanoparticles
- NAC, N-acetyl-l-cysteine
- NLCs, nanostructured lipid carriers
- Oral delivery
- PAA, polyacrylic acid
- PBPK, physiologically based pharmacokinetics
- PCA, principal component analysis
- PCL, polycarprolacton
- PGA, poly-γ-glutamic acid
- PLA, poly(latic acid)
- PLGA, poly(lactic-co-glycolic acid)
- PPs, proteins and peptides
- PVA, poly vinyl alcohol
- Peptides
- Permeation enhancer
- Proteins
- RGD, Arg-Gly-Asp
- RTILs, room temperature ionic liquids
- SAR, structure–activity relationship
- SDC, sodium deoxycholate
- SGC, sodium glycocholate
- SGF, simulated gastric fluids
- SIF, simulated intestinal fluids
- SLNs, solid lipid nanoparticles
- SNAC, sodium N-[8-(2-hydroxybenzoyl)amino]caprylate
- SNEDDS, self-nanoemulsifying drug delivery systems
- STC, sodium taurocholate
- Stability
- TAT, trans-activating transcriptional peptide
- TMC, N-trimethyl chitosan
- Tf, transferrin
- TfR, transferrin receptors
- UC, ulcerative colitis
- UEA1, ulex europaeus agglutinin 1
- VB12, vitamin B12
- WGA, wheat germ agglutinin
- pHPMA, N-(2-hydroxypropyl)methacrylamide
- pI, isoelectric point
- sCT, salmon calcitonin
- sc, subcutaneous
Collapse
Affiliation(s)
- Quangang Zhu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Pijush Kumar Paul
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Department of Pharmacy, Gono Bishwabidyalay (University), Mirzanagar Savar, Dhaka 1344, Bangladesh
| | - Yi Lu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wei Wu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jianping Qi
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
34
|
Lai X, Tang J, ElSayed MEH. Recent advances in proteolytic stability for peptide, protein, and antibody drug discovery. Expert Opin Drug Discov 2021; 16:1467-1482. [PMID: 34187273 DOI: 10.1080/17460441.2021.1942837] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: To discover and develop a peptide, protein, or antibody into a drug requires overcoming multiple challenges to obtain desired properties. Proteolytic stability is one of the challenges and deserves a focused investigation.Areas covered: This review concentrates on improving proteolytic stability by engineering the amino acids around the cleavage sites of a liable peptide, protein, or antibody. Peptidases are discussed on three levels including all peptidases in databases, mixtures based on organ and tissue types, and individual peptidases. The technique to identify cleavage sites is spotlighted on mass spectrometry-based approaches such as MALDI-TOF and LC-MS. For sequence engineering, the replacements that have been commonly applied with a higher chance of success are highlighted at the beginning, while the rarely used and more complicated replacements are discussed later. Although a one-size-fits-all approach does not exist to apply to different projects, this review provides a 3-step strategy for effectively and efficiently conducting the proteolytic stability experiments to achieve the eventual goal of improving the stability by engineering the molecule itself.Expert opinion: Improving the proteolytic stability is a spiraling up process sequenced by testing and engineering. There are many ways to engineer amino acids, but the choice must consider the cost and properties affected by the changes of the amino acids.
Collapse
Affiliation(s)
- Xianyin Lai
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jason Tang
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mohamed E H ElSayed
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
35
|
Modi NB, Cheng X, Mattheakis L, Hwang CC, Nawabi R, Liu D, Gupta S. Single- and Multiple-Dose Pharmacokinetics and Pharmacodynamics of PN-943, a Gastrointestinal-Restricted Oral Peptide Antagonist of α4β7, in Healthy Volunteers. Clin Pharmacol Drug Dev 2021; 10:1263-1278. [PMID: 33942566 PMCID: PMC8597174 DOI: 10.1002/cpdd.946] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/07/2021] [Indexed: 12/19/2022]
Abstract
PN‐943 is an orally stable, gastrointestinal‐restricted peptide that binds specifically to α4ß7 integrin on leukocytes, blocking leukocyte trafficking to and activation in the gut, inhibiting colon inflammation and reducing signs and symptoms of active ulcerative colitis. Two pharmacokinetic/pharmacodynamic studies were conducted in healthy volunteers. Study 1 was a first‐in‐human study with 40 male subjects receiving PN‐943, 100 to 1400 mg or placebo, as single doses and 57 male subjects receiving PN‐943, 100 to 1000 mg or placebo, as multiple doses. Study 2 was a randomized, crossover study comparing multiple doses of 450‐mg PN‐943 twice daily as a liquid solution and as an immediate‐release tablet in 10 subjects. No subjects discontinued due to treatment‐emergent adverse events. Consistent with the gastrointestinal‐restricted nature of the peptide, systemic exposure was minimal; there was an approximate dose‐proportional increase in area under the plasma concentration–time curve. There was minimal accumulation with once‐daily dosing and an absence of time‐dependent changes in pharmacokinetics. Administration of PN‐943 after a high‐fat meal reduced peak plasma concentration and area under the plasma concentration–time curve. There was minimal (<0.1%) urinary excretion of intact drug, and there was a dose‐related increase in fecal excretion of intact PN‐943. Dose‐dependent increases in blood receptor occupancy and reduction in blood receptor expression were observed, supporting target engagement. Twice‐daily dosing resulted in sustained receptor occupancy with low plasma fluctuations (143%). PN‐943 was generally well tolerated following single and multiple oral doses with low systemic exposure. Twice‐daily dosing resulted in sustained pharmacokinetics and pharmacodynamics, supporting further investigation in efficacy studies.
Collapse
Affiliation(s)
- Nishit B Modi
- Protagonist Therapeutics, Inc, Newark, California, USA
| | - Xiaoli Cheng
- Protagonist Therapeutics, Inc, Newark, California, USA
| | | | | | - Roya Nawabi
- Protagonist Therapeutics, Inc, Newark, California, USA
| | - David Liu
- Protagonist Therapeutics, Inc, Newark, California, USA
| | - Suneel Gupta
- Protagonist Therapeutics, Inc, Newark, California, USA
| |
Collapse
|
36
|
Li W, Zhu X, Zhou X, Wang X, Zhai W, Li B, Du J, Li G, Sui X, Wu Y, Zhai M, Qi Y, Chen G, Gao Y. An orally available PD-1/PD-L1 blocking peptide OPBP-1-loaded trimethyl chitosan hydrogel for cancer immunotherapy. J Control Release 2021; 334:376-388. [PMID: 33940058 DOI: 10.1016/j.jconrel.2021.04.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/13/2021] [Accepted: 04/29/2021] [Indexed: 01/06/2023]
Abstract
Blockade of the immune checkpoint PD-1/PD-L1 with monoclonal antibodies demonstrated unprecedented clinical efficacies in many cancers. But the orally available low molecular weight inhibitors remain infancy. Compared to small molecules, peptide exhibits better selectivity and fewer side effects, but poor half-life and a big challenge to be orally administrated. Here, we developed a proteolysis-resistant D peptide OPBP-1 (Oral PD-L1 Binding Peptide 1) which could selectively bind PD-L1, significantly block PD-1/PD-L1 interaction and enhance IFN-γ (interferon γ) secretion from CD8+ T cells in human PBMCs (Peripheral blood mononuclear cells). OPBP-1 could significantly inhibit tumor growth in murine colorectal CT26 and melanoma B16-OVA models at a relatively low dose of 0.5 mg/kg, with enhancing the infiltration and function of CD8+ T cells. More interestingly, oral delivery of OPBP-1 loaded TMC (N, N, N-trimethyl chitosan) hydrogel (OPBP-1@TMC) showed promising OPBP-1 oral bioavailability (52.8%) and prolonged half-life (14.55 h) in rats, and also significantly inhibited tumor growth in CT26 model. In conclusion, we discovered and optimized a PD-1/PD-L1 blocking peptide OPBP-1, and subsequently loaded into a TMC based hydrogel oral delivery system, in order to maximally elevate the oral bioavailability of the peptide drug and effectively inhibit tumor growth. These results opened up a new prospect for oral drug development in cancer immunotherapy.
Collapse
Affiliation(s)
- Wanqiong Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China; School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xueqin Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiuman Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaoxi Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Bingyu Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jiangfeng Du
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Guodong Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xinghua Sui
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Mingxia Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Guanyu Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China; School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
37
|
Ren T, Zheng X, Bai R, Yang Y, Jian L. Utilization of PLGA nanoparticles in yeast cell wall particle system for oral targeted delivery of exenatide to improve its hypoglycemic efficacy. Int J Pharm 2021; 601:120583. [PMID: 33839225 DOI: 10.1016/j.ijpharm.2021.120583] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/14/2021] [Accepted: 04/04/2021] [Indexed: 11/19/2022]
Abstract
Oral delivery of exenatide (EXE), a high-efficiency therapeutic peptide, is urgently needed for long-term treatment of diabetes. In this study, a polylactide-co-glycoside (PLGA) nanoparticles (NPs) in yeast cell wall particle (YCWP) system was built to improve the intestinal absorption of EXE by efficient protection of EXE against gastrointestinal degradation and intestinal phagocytic cell targeted delivery. The EXE-loaded PLGA NPs were prepared by a double emulsion solvent diffusion method and exhibited a uniformly spherical appearance, a nano size (92.4 ± 4.6 nm) and a positive surface charge (+32.3 ± 3.8 mV). And then, the NPs were successfully loaded into the YCWPs by a solvent hydration - lyophilization cycle method to obtain the EXE-PLGA NPs @YCWPs, which was verified by scanning electron microscope and confocal laser scanning microscopy. An obvious sustained drug release and a reduced burst release were achieved by this nano-in-micro carrier. Moreover, the gastrointestinal stability of EXE in PLGA NPs @YCWPs was significantly higher than that in PLGA NPs in the simulated gastrointestinal environment, which were useful in enhancing the intestinal absorption of EXE. In biodistribution study, the EXE-PLGA NPs @YCWPs could quickly reached the root of the villi, and even partly entered the inner of the villi, especially in ileum and Peyer's patches. In vitro cell evaluation demonstrated an efficient β-glucan receptor mediated endocytosis and transport of EXE-PLGA NPs @YCWPs by the macrophage RAW 264.7 cells, suggesting a potential intestinal macrophage targeted absorptive pathway. The in vivo pharmacokinetic study showed a preferred hypoglycemic effect and an increased pharmacological availability (13.7 ± 4.1%) after oral administration of the EXE-PLGA NPs @YCWPs. It is believed that the PLGA nanoparticles in YCWP system could become an efficient strategy to orally deliver therapeutic peptide drugs.
Collapse
Affiliation(s)
- Tianyang Ren
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Xuehua Zheng
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Ruixue Bai
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Yuehui Yang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, PR China.
| | - Lingyan Jian
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
38
|
Losacco GL, DaSilva JO, Liu J, Regalado EL, Veuthey JL, Guillarme D. Expanding the range of sub/supercritical fluid chromatography: Advantageous use of methanesulfonic acid in water-rich modifiers for peptide analysis. J Chromatogr A 2021; 1642:462048. [PMID: 33744606 DOI: 10.1016/j.chroma.2021.462048] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 11/30/2022]
Abstract
The aim of this work was to expand the applicability range of UHPSFC to series of synthetic and commercialized peptides. Initially, a screening of different column chemistries available for UHPSFC analysis was performed, in combination with additives of either basic or acidic nature. The combination of an acidic additive (13 mM TFA) with a basic stationary phase (Torus DEA and 2-PIC) was found to be the best for a series of six synthetic peptides possessing either acidic, neutral or basic isoelectric points. Secondly, methanesulfonic acid (MSA) was evaluated as a potential replacement for TFA. Due to its stronger acidity, MSA gave better performance than TFA at the same concentration level. Furthermore, the use of reduced percentages of MSA, such as 8 mM, yielded similar results to those observed with 15 mM of MSA. The optimized UHPSFC method was, then, used to compare the performance of UHPSFC against RP-UHPLC for peptides with different pI and with increasing peptide chain length. UHPSFC was found to give a slightly better separation of the peptides according to their pI values, in few cases orthogonal to that observed in UHPLC. On the other hand, UHPSFC produced a much better separation of peptides with an increased amino acidic chain compared to UHPLC. Subsequently, UHPSFC-MS was systematically compared to UHPLC-MS using a set of linear and cyclic peptides commercially available. The optimized UHPSFC method was able to generate at least similar, and in some cases even better performance to UHPLC with the advantage of providing complementary information to that given by UHPLC analysis. Finally, the analytical UHPSFC method was transferred to a semipreparative scale using a proprietary cyclic peptide, demonstrating excellent purity and high yield in less than 15 min.
Collapse
Affiliation(s)
- Gioacchino Luca Losacco
- School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel-Servet 1, 1211 Geneva 4, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - Jimmy Oliviera DaSilva
- Analytical Research and Development, MRL, Merck & Co, Inc., 126 E. Lincoln Ave, Rahway, NJ 07065, United States
| | - Jinchu Liu
- Analytical Research and Development, MRL, Merck & Co, Inc., 126 E. Lincoln Ave, Rahway, NJ 07065, United States
| | - Erik L Regalado
- Analytical Research and Development, MRL, Merck & Co, Inc., 126 E. Lincoln Ave, Rahway, NJ 07065, United States
| | - Jean-Luc Veuthey
- School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel-Servet 1, 1211 Geneva 4, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - Davy Guillarme
- School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel-Servet 1, 1211 Geneva 4, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel-Servet 1, 1211 Geneva 4, Switzerland.
| |
Collapse
|
39
|
Tee YN, Kumar PV, Maki MAA, Elumalai M, Rahman SAKMEH, Cheah SC. Mucoadhesive Low Molecular Chitosan Complexes to Protect rHuKGF from Proteolysis: In-vitro Characterization and FHs 74 Int Cell Proliferation Studies. Curr Pharm Biotechnol 2021; 22:969-982. [PMID: 33342408 DOI: 10.2174/1389201021666201218124450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/15/2020] [Accepted: 10/24/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Recombinant Keratinocyte Growth Factor (rHuKGF) is a therapeutic protein used widely in oral mucositis after chemotherapy in various cancers, stimulating lung morphogenesis and gastrointestinal tract cell proliferation. In this research study, chitosan-rHuKGF polymeric complex was implemented to improve the stability of rHuKGF and used as rejuvenation therapy for the treatment of oral mucositis in cancer patients. OBJECTIVE Complexation of rHuKGF with mucoadhesive low molecular weight chitosan to protect rHuKGF from proteolysis and investigate the effect of chitosan-rHuKGF complex on the proliferation rate of FHs 74 Int cells. METHODS The interaction between chitosan and rHuKGF was studied by molecular docking. Malvern ZetaSizer Nano Zs and Fourier-Transform Infrared spectroscopy (FTIR) tests were carried out to characterize the chitosan-rHuKGF complex. In addition, SDS-PAGE was performed to investigate the interaction between chitosan-rHuKGF complex and pepsin. The effect of chitosan-rHuKGF complex on the proliferation rate of FHs 74 Int cells was studied by MTT assay. RESULTS Chitosan-rHuKGF complex was formed through the hydrogen bonding proven by the docking studies. A stable chitosan-rHuKGF complex was formed at pH 4.5 and was protected from proteolysis and assessed by SDS PAGE. According to the MTT assay results, chitosan-rHuKGF complex increased the cell proliferation rate of FHs 74 Int cells. CONCLUSION The developed complex improved the stability and the biological function of rHuKGF.
Collapse
Affiliation(s)
- Yi N Tee
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras 56000 Kuala Lumpur, Malaysia
| | - Palanirajan V Kumar
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras 56000 Kuala Lumpur, Malaysia
| | - Marwan A A Maki
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras 56000 Kuala Lumpur, Malaysia
| | - Manogaran Elumalai
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras 56000 Kuala Lumpur, Malaysia
| | - Shiek A K M E H Rahman
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras 56000 Kuala Lumpur, Malaysia
| | - Shiau-Chuen Cheah
- Faculty of Medicine & Health Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras 56000 Kuala Lumpur, Malaysia
| |
Collapse
|
40
|
Water/pH dual responsive in situ calcium supplement collaborates simvastatin for osteoblast promotion mediated osteoporosis therapy via oral medication. J Control Release 2020; 329:121-135. [PMID: 33279604 DOI: 10.1016/j.jconrel.2020.11.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/22/2020] [Accepted: 11/29/2020] [Indexed: 12/20/2022]
Abstract
Calcium supplement is the most commonly adopted treatment for osteoporosis but usually requires high dose and frequency. The modality of calcium supplement is therefore overlooked by current nanomedicine-based osteoporosis therapies without proper oral formulations. Herein, we proposed a tetracycline (Tc) modified and monostearin (MS) coated amorphous calcium carbonate (ACC) platform (TMA) as oral bone targeted and osteoporosis microenvironment (water/pH) responsive carrier for in situ calcium supplement. Moreover, current osteoporosis therapies also fall short of finding suitable molecular target and effective therapeutic regimen to further increase the therapeutic efficacy over available treatment means. As a result, the simvastatin (Sim) was loaded into TMA to construct drug delivery system (TMA/Sim) capable of synergistically activating the bone morphogenetic proteins (BMPs)-Smad pathway to provide a novel therapeutic regimen for osteoblast promotion mediated osteoporosis therapy. Our results revealed that optimized TMA showed high accessibility and oral availability with targeted drug delivery to bone tissue. Most importantly, benefit from the effective in situ calcium supplement and targeted Sim delivery, this therapeutic regime (TMA/Sim) achieved better synergetic effects than conventional combination strategies with promising osteoporosis reversion performance under low calcium dosage (1/10 of commercial calcium carbonate tablet) and significantly attenuated side effects.
Collapse
|
41
|
Hsu BB, Plant IN, Lyon L, Anastassacos FM, Way JC, Silver PA. In situ reprogramming of gut bacteria by oral delivery. Nat Commun 2020; 11:5030. [PMID: 33024097 PMCID: PMC7538559 DOI: 10.1038/s41467-020-18614-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
Abundant links between the gut microbiota and human health indicate that modification of bacterial function could be a powerful therapeutic strategy. The inaccessibility of the gut and inter-connections between gut bacteria and the host make it difficult to precisely target bacterial functions without disrupting the microbiota and/or host physiology. Herein we describe a multidisciplinary approach to modulate the expression of a specific bacterial gene within the gut by oral administration. We demonstrate that an engineered temperate phage λ expressing a programmable dCas9 represses a targeted E. coli gene in the mammalian gut. To facilitate phage administration while minimizing disruption to host processes, we develop an aqueous-based encapsulation formulation with a microbiota-based release mechanism and show that it facilitates oral delivery of phage in vivo. Finally we combine these technologies and show that bacterial gene expression in the mammalian gut can be precisely modified in situ with a single oral dose.
Collapse
Affiliation(s)
- Bryan B Hsu
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| | - Isaac N Plant
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Lorena Lyon
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Frances M Anastassacos
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jeffrey C Way
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Pamela A Silver
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
42
|
De Leon Rodriguez LM, Hemar Y. Prospecting the applications and discovery of peptide hydrogels in food. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2020.07.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
43
|
Abeer MM, Rewatkar P, Qu Z, Talekar M, Kleitz F, Schmid R, Lindén M, Kumeria T, Popat A. Silica nanoparticles: A promising platform for enhanced oral delivery of macromolecules. J Control Release 2020; 326:544-555. [DOI: 10.1016/j.jconrel.2020.07.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/15/2022]
|
44
|
Sharkey SJ, Harnedy-Rothwell PA, Allsopp PJ, Hollywood LE, FitzGerald RJ, O'Harte FPM. A Narrative Review of the Anti-Hyperglycemic and Satiating Effects of Fish Protein Hydrolysates and Their Bioactive Peptides. Mol Nutr Food Res 2020; 64:e2000403. [PMID: 32939966 DOI: 10.1002/mnfr.202000403] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Prevalence of type 2 diabetes and overweight/obesity are increasing globally. Food supplementation as a preventative option has become an attractive option in comparison to increased pharmacotherapy dependency. Hydrolysates of fish processing waste and by-products have become particularly interesting in a climate of increased food wastage awareness and are rapidly gaining traction in food research. This review summarizes the available research so far on the potential effect of these hydrolysates on diabetes and appetite suppression. Scopus and Web of Science are searched using eight keywords (fish, hydrolysate, peptides, satiating, insulinotropic, incretin, anti-obesity, DPP-4 [dipeptidylpeptidase-4/IV]) returning a total of 2549 results. Following exclusion criteria (repeated appearances, non-fish marine sources [e.g., macroalgae], and irrelevant bioactivities [e.g., immunomodulatory, anti-thrombotic]), 44 relevant publications are included in this review. Stimulation of hormone secretion, regulation of glucose uptake, anorexigenic potential, identified mechanisms of action, and research conducted on the most potent bioactive peptides identified within these hydrolysates are all specifically addressed. Results of this review conclude that despite wide methodological variation between studies, there is significant potential for the application of fish protein hydrolysates in the management of bodyweight and hyperglycemia.
Collapse
Affiliation(s)
- Shaun J Sharkey
- School of Biomedical Sciences, Ulster University, Cromore Road, Co. Derry, Northern Ireland, Coleraine, BT52 1SA, UK
| | | | - Philip J Allsopp
- School of Biomedical Sciences, Ulster University, Cromore Road, Co. Derry, Northern Ireland, Coleraine, BT52 1SA, UK
| | - Lynsey E Hollywood
- Department of Hospitality and Tourism Management, Ulster University Business School, Ulster University, Co. Derry, Northern Ireland, Coleraine, BT52 1SA, UK
| | - Richard J FitzGerald
- Department of Biological Sciences, University of Limerick, Castletroy, Limerick, V94 T9PX, Ireland
| | - Finbarr P M O'Harte
- School of Biomedical Sciences, Ulster University, Cromore Road, Co. Derry, Northern Ireland, Coleraine, BT52 1SA, UK
| |
Collapse
|
45
|
Liu J, Leng P, Liu Y. Oral drug delivery with nanoparticles into the gastrointestinal mucosa. Fundam Clin Pharmacol 2020; 35:86-96. [PMID: 32749731 DOI: 10.1111/fcp.12594] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 12/16/2022]
Abstract
The oral route of protein and peptide drugs has been a popular method of drug delivery in recent years, although it is often a challenge to achieve effective drug release and minimize the barrier functions of the gastrointestinal tract. Gastrointestinal mucosa can capture and remove harmful substances; similarly, it can limit the absorption of drugs. Many drugs are effectively captured by the mucus and rapidly removed, making it difficult to control the release of drugs in the gastrointestinal tract. The use of drug carrier systems can overcome the mucosal barrier and significantly improve bioavailability. Nanoparticle drug carriers can protect the drug from degradation, transporting it to a predetermined location in the gastrointestinal tract to achieve more efficient and sustained drug delivery. It is becoming clearer that the characteristics of nanoparticles, such as particle size, charge, and hydrophobicity, are related to permeability of the mucosal barrier. This review focuses on the latest research progress of nanoparticles to penetrate the mucosal barrier, including the delivery methods of nanoparticles on the surface of gastrointestinal mucosa, and aims to summarize how successful oral nanoparticle delivery systems can overcome this biological barrier in the human body. In addition, the in vitro model based on gastrointestinal mucus is an important tool for drug research and development. Here, we discuss different types of drug delivery systems and their advantages and disadvantages in design and potential applications. Similarly, we reviewed and summarized various methods for evaluating oral nanoparticles in in vitro and in vivo models.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China
| | - Ping Leng
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China
| | - Yujun Liu
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China
| |
Collapse
|
46
|
Mertins O, Mathews PD, Angelova A. Advances in the Design of pH-Sensitive Cubosome Liquid Crystalline Nanocarriers for Drug Delivery Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E963. [PMID: 32443582 PMCID: PMC7281514 DOI: 10.3390/nano10050963] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 04/29/2020] [Accepted: 05/03/2020] [Indexed: 01/10/2023]
Abstract
Nanostructure bicontinuous cubic phase self-assembled materials are receiving expanding applications as biocompatible delivery systems in various therapeutic fields. The functionalization of cubosome, spongosome, hexosome and liposome nanocarriers by pH-sensitive lipids and/or pH-sensitive polymer shells offers new opportunities for oral and topical drug delivery towards a new generation of cancer therapies. The electrochemical behavior of drug compounds may favor pH-triggered drug release as well. Here, we highlight recent investigations, which explore the phase behavior of mixed nonlamellar lipid/fatty acid or phospholipid systems for the design of pH-responsive and mucoadhesive drug delivery systems with sustained-release properties. X-ray diffraction and small-angle X-ray scattering (SAXS) techniques are widely used in the development of innovative delivery assemblies through detailed structural analyses of multiple amphiphilic compositions from the lipid/co-lipid/water phase diagrams. pH-responsive nanoscale materials and nanoparticles are required for challenging therapeutic applications such as oral delivery of therapeutic proteins and peptides as well as of poorly water-soluble substances. Perspective nanomedicine developments with smart cubosome nanocarriers may exploit compositions elaborated to overcome the intestinal obstacles, dual-drug loaded pH-sensitive liquid crystalline architectures aiming at enhanced therapeutic efficacy, as well as composite (lipid/polyelectrolyte) types of mucoadhesive controlled release colloidal cubosomal formulations for the improvement of the drugs' bioavailability.
Collapse
Affiliation(s)
- Omar Mertins
- Institut Galien Paris-Saclay UMR8612, Université Paris-Saclay, CNRS, F-92296 Châtenay-Malabry, France;
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), Sao Paulo 04023-062, Brazil;
| | - Patrick D. Mathews
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), Sao Paulo 04023-062, Brazil;
- Muséum National d’Histoire Naturelle, Sorbonne Université, CP 26, 75231 Paris, France
| | - Angelina Angelova
- Institut Galien Paris-Saclay UMR8612, Université Paris-Saclay, CNRS, F-92296 Châtenay-Malabry, France;
| |
Collapse
|
47
|
Yin M, Song Y, Guo S, Zhang X, Sun K, Li Y, Shi Y. Intelligent Escape System for the Oral Delivery of Liraglutide: A Perfect Match for Gastrointestinal Barriers. Mol Pharm 2020; 17:1899-1909. [PMID: 32267705 DOI: 10.1021/acs.molpharmaceut.9b01307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Epithelial cells are known to impede the oral delivery of polypeptides, and the accumulation of mucus and regular dynamic renewal also significantly impede drug absorption. In this work, we prepared a core-shell (COS) nanosystem using poly-N-(2-hydroxypropyl)methacrylamide (pHPMA)/chitosan (CTS). Liraglutide (NN2211) was isolated from the gastrointestinal environment and smoothly passes through the mucous layer. CSKSSDYQC (CSK) peptide and hemagglutinin-2 (HA2) were introduced into the COS nanosystem to establish a complete path from the oral cavity to the epithelial basal side. The fate of nanocapsules in vivo was studied by fluorescence detection. The results showed that the nanocapsules escaped smoothly from the mucus. Taking into account the characteristics of CSK targeting goblet cells, we conducted cell-level studies, and the results showed that after the modification of CSK and pHPMA, more nanocapsules entered the cells. In vitro and in vivo evaluation results showed that the system successfully established a complete path from mucus to epithelial cells by responding to the gastrointestinal environment multiple times.
Collapse
Affiliation(s)
- Miaomiao Yin
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P. R. China
| | - Yina Song
- State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai 264003, P. R. China
| | - Shiqi Guo
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P. R. China
| | - Xuemei Zhang
- State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai 264003, P. R. China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P. R. China.,State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai 264003, P. R. China
| | - Youxin Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P. R. China.,State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai 264003, P. R. China
| | - Yanan Shi
- School of Life Science, Yantai University, Yantai 264005, P. R. China
| |
Collapse
|
48
|
Knerr PJ, Mowery SA, Finan B, Perez-Tilve D, Tschöp MH, DiMarchi RD. Selection and progression of unimolecular agonists at the GIP, GLP-1, and glucagon receptors as drug candidates. Peptides 2020; 125:170225. [PMID: 31786282 DOI: 10.1016/j.peptides.2019.170225] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
Abstract
The continued global growth in the prevalence of obesity coupled with the limited number of efficacious and safe treatment options elevates the importance of innovative pharmaceutical approaches. Combinatorial strategies that harness the metabolic benefits of multiple hormonal mechanisms have emerged at the preclinical and more recently clinical stages of drug development. A priority has been anti-obesity unimolecular peptides that function as balanced, high potency poly-agonists at two or all the cellular receptors for the endocrine hormones glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), and glucagon. This report reviews recent progress in this area, with emphasis on what the initial clinical results demonstrate and what remains to be addressed.
Collapse
Affiliation(s)
- Patrick J Knerr
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | | | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Diego Perez-Tilve
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Garching, Germany
| | - Richard D DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA; Department of Chemistry, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
49
|
Dahlgren D, Sjöblom M, Hedeland M, Lennernäs H. The In Vivo Effect of Transcellular Permeation Enhancers on the Intestinal Permeability of Two Peptide Drugs Enalaprilat and Hexarelin. Pharmaceutics 2020; 12:pharmaceutics12020099. [PMID: 31991924 PMCID: PMC7076382 DOI: 10.3390/pharmaceutics12020099] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/14/2022] Open
Abstract
Permeation enhancers like sodium dodecyl sulfate (SDS) and caprate increase the intestinal permeability of small model peptide compounds, such as enalaprilat (349 Da). However, their effects remain to be investigated for larger low-permeability peptide drugs, such as hexarelin (887 Da). The objective of this single-pass perfusion study in rat was to investigate the effect of SDS at 5 mg/mL and of caprate administered at different luminal concentrations (5, 10, and 20 mg/mL) and pH (6.5 and 7.4). The small intestinal permeability of enalaprilat increased by 8- and 9-fold with SDS at 5 mg/mL and with caprate at 10 and 20 mg/mL but only at pH 7.4, where the free dissolved caprate concentration is higher than at pH 6.5 (5 vs. 2 mg/mL). Neither SDS nor caprate at any of the investigated luminal concentrations enhanced absorption of the larger peptide hexarelin. These results show that caprate requires doses above its saturation concentration (a reservoir suspension) to enhance absorption, most likely because dissolved caprate itself is rapidly absorbed. The absent effect on hexarelin may partly explain why the use of permeation enhancers for enabling oral peptide delivery has largely failed to evolve from in vitro evaluations into approved oral products. It is obvious that more innovative and effective drug delivery strategies are needed for this class of drugs.
Collapse
Affiliation(s)
- David Dahlgren
- Department of Pharmacy, Uppsala University, 751 23 Uppsala, Sweden;
| | - Markus Sjöblom
- Department of Neuroscience, Uppsala University, 751 23 Uppsala, Sweden;
| | - Mikael Hedeland
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden;
- National Veterinary Institute (SVA), 751 89 Uppsala, Sweden
| | - Hans Lennernäs
- Department of Pharmacy, Uppsala University, 751 23 Uppsala, Sweden;
- Correspondence: ; Tel.: +46-18-471-4317
| |
Collapse
|
50
|
Praveschotinunt P, Duraj-Thatte AM, Gelfat I, Bahl F, Chou DB, Joshi NS. Engineered E. coli Nissle 1917 for the delivery of matrix-tethered therapeutic domains to the gut. Nat Commun 2019; 10:5580. [PMID: 31811125 PMCID: PMC6898321 DOI: 10.1038/s41467-019-13336-6] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/29/2019] [Indexed: 02/06/2023] Open
Abstract
Mucosal healing plays a critical role in combatting the effects of inflammatory bowel disease, fistulae and ulcers. While most treatments for such diseases focus on systemically delivered anti-inflammatory drugs, often leading to detrimental side effects, mucosal healing agents that target the gut epithelium are underexplored. We genetically engineer Escherichia coli Nissle 1917 (EcN) to create fibrous matrices that promote gut epithelial integrity in situ. These matrices consist of curli nanofibers displaying trefoil factors (TFFs), known to promote intestinal barrier function and epithelial restitution. We confirm that engineered EcN can secrete the curli-fused TFFs in vitro and in vivo, and is non-pathogenic. We observe enhanced protective effects of engineered EcN against dextran sodium sulfate-induced colitis in mice, associated with mucosal healing and immunomodulation. This work lays a foundation for the development of a platform in which the in situ production of therapeutic protein matrices from beneficial bacteria can be exploited.
Collapse
Affiliation(s)
- Pichet Praveschotinunt
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Anna M Duraj-Thatte
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Ilia Gelfat
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Franziska Bahl
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Faculty of Biology, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - David B Chou
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Neel S Joshi
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| |
Collapse
|