1
|
Yoon BK, Jackman JA. Medium-chain fatty acids and monoglycerides: Nanoarchitectonics-based insights into molecular self-assembly, membrane interactions, and applications. Adv Colloid Interface Sci 2025; 340:103465. [PMID: 40056558 DOI: 10.1016/j.cis.2025.103465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/10/2025]
Abstract
Medium-chain fatty acids (FAs) and monoglycerides (MGs) with saturated 6- to 12‑carbon long tails are single-chain lipid amphiphiles that demonstrate significant application merits. Key examples include their antimicrobial activity against antibiotic-resistant bacteria and emerging viral threats as well as innovations in oral pharmaceutics and biorenewable chemical production. These diverse functionalities are enabled by FA and MG self-assembly and their interactions with biological membranes. However, an integrated viewpoint connecting interfacial science principles to the broader application scope remains lacking. The objective of this review is to cover the latest progress in medium-chain FA and MG research and to build connections between molecular self-assembly, membrane interactions, and applications. By taking a bottom-up nanoarchitectonics perspective, we first examine molecular self-assembly principles, including ionization properties and formation of colloidal nanostructures such as micelles and vesicles. We then discuss membrane interaction concepts and experimental findings that illustrate how medium-chain FAs and MGs distinctly interact with phospholipid membranes. Based on this foundation, we highlight cutting-edge applications in medicine, agriculture, drug delivery, and sustainability, linking these advances to interfacial science concepts. In addition, we emphasize the growing convergence of experimental, theoretical, and computational approaches and offer a forward-looking perspective on future research needs and application opportunities.
Collapse
Affiliation(s)
- Bo Kyeong Yoon
- School of Biomedical Engineering, Chonnam National University, Yeosu, Republic of Korea
| | - Joshua A Jackman
- School of Chemical Engineering and Translational Nanobioscience Research Center, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
2
|
Bernaldez M, Kang C, Stamatis SD, Rose JP, Sun R. The Impact of Permeation Enhancers on Transcellular Permeation of Small Molecule Drugs. J Phys Chem B 2025. [PMID: 40378260 DOI: 10.1021/acs.jpcb.5c00953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Passive permeation through an epithelial membrane may be enhanced by using a class of amphiphilic molecules known as permeation enhancers (PEs). PEs have been studied in clinical trials and used in coformulations with peptides and small molecule drugs, and yet, an understanding of the permeant-PE interactions leaves much to be desired. This manuscript uses all-atom molecular dynamics (MD) simulations to showcase the effects of sodium caprate (C10) and salcaprozate sodium (SNAC), two commonly applied PEs, on membrane properties and the free energy profiles of five small molecule drugs (mannitol, atenolol, ketoprofen, decanedecaol, mucic acid). Our results show that both C10 and SNAC make the lipid molecules pack more densely, but C10 increases the lipid lateral diffusivity while SNAC decreases it. The change in the lipid order parameter also shows both PEs increasing the order near the lipid heads, possibly due to the dense packing in the membrane. A decrease in the central barrier of the permeation free energy was observed by embedding PEs into a lipid bilayer and SNAC is more efficient in doing so than C10. Neither SNAC nor C10 has a large impact on the diffusion coefficient of the small molecules. The analysis of the MD simulations revealed that PEs make the membrane tail region more hydrophilic by forming hydrogen bonds with small molecule drugs, i.e., decreasing the central barrier of the permeation free energy. While this study was only limited to small molecule drugs, this lays the groundwork for future studies to which the effects of the PEs in the permeation of macromolecules and peptides may be observed.
Collapse
Affiliation(s)
- Mabel Bernaldez
- Department of Chemistry, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| | - Christopher Kang
- Department of Chemistry, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| | - Stephen D Stamatis
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - John P Rose
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Rui Sun
- Department of Chemistry, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| |
Collapse
|
3
|
Niu Z, La Zara D, Blaabjerg L, Pessi J, Raptis K, Toftlev A, Sauter M, Christophersen P, Bardonnet PL, Andersson V, Wu JX, Brandt M, Fan L, Wang Z, Hubálek F, Wahlund PO, Norrman M, Breusova K, Hjaltason MS, Mortensen NR, Bardtrum L, Nissen B, Naelapää K, Sassene PJ. Combining SNAC and C10 in oral tablet formulations for gastric peptide delivery: A preclinical and clinical study. J Control Release 2025; 378:92-102. [PMID: 39645088 DOI: 10.1016/j.jconrel.2024.11.078] [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: 04/15/2024] [Revised: 10/03/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Current oral formulations of macromolecules including peptides typically rely on single permeation enhancer (PE) to promote absorption and thus bioavailability. In this work, we combined two PEs, namely sodium N-[8-(2-hydroxybenzoyl) amino] caprylate (SNAC) and sodium caprate (C10), in one tablet formulation to potentially gain a synergistic effect for enhanced gastric absorption of a GLP-1 analogue and a PCSK9 inhibitor. Permeability tests on a gastric organoids-based cell model showed that the combination of SNAC and C10 can significantly improve peptide permeability compared to either SNAC or C10 alone. Tablet formulations were then designed, adjusting the total PE amount, relative ratio between SNAC and C10, and the peptide dose. To facilitate drug and PE release, a diluent was added. Upon oral administration in beagle dogs, the lead formulations made of SNAC/C10/diluent demonstrated higher bioavailability than either SNAC, SNAC/diluent and C10/diluent formulations for both peptides. Finally, the SNAC/C10/diluent formulation with PCSK9 inhibitor was tested in human, where it displayed similar bioavailability to the SNAC/diluent reference, thereby suggesting a low translatability between pre-clinical and clinical data when C10 was involved. This may be attributed to the difference in physiology, gastric pH environment as well as C10 concentration and colloidal form in the gastric lumen between dogs and humans. Hence, additional studies are needed for a better understanding of the clinical translation of C10-based peptide formulations.
Collapse
Affiliation(s)
- Zhigao Niu
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark.
| | - Damiano La Zara
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | - Lasse Blaabjerg
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | - Jenni Pessi
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | | | - Anders Toftlev
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | - Max Sauter
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | | | | | | | - Jian Xiong Wu
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | - Matthäus Brandt
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | - Li Fan
- Novo Nordisk Research Centre China, 102206 Beijing, China
| | - Zhuoran Wang
- Novo Nordisk Research Centre China, 102206 Beijing, China
| | - Franta Hubálek
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | - Per-Olof Wahlund
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | - Mathias Norrman
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | | | | | | | - Lars Bardtrum
- Novo Nordisk A/S, Biostatistics, 2860 Søborg, Denmark
| | - Birgitte Nissen
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | - Kaisa Naelapää
- Novo Nordisk A/S, Global Research Technologies, 2760 Måløv, Denmark
| | | |
Collapse
|
4
|
Ling J, Schroder R, Wuelfing WP, Higgins J, Kesisoglou F, Templeton AC, Su Y. Molecular Investigation of SNAC as an Oral Peptide Permeation Enhancer in Lipid Membranes via Solid-State NMR. Mol Pharm 2025; 22:459-473. [PMID: 39690106 DOI: 10.1021/acs.molpharmaceut.4c01061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Oral peptide therapeutics are increasingly favored in the pharmaceutical industry for their ease of use and better patient adherence. However, they face challenges with poor oral bioavailability due to their high molecular weight and surface polarity. Permeation enhancers (PEs) like salcaprozate sodium (SNAC) have shown promise in clinical trials, achieving about 1% bioavailability. One proposed mechanism for enhancing permeation is membrane perturbation or fluidization, though direct experimental proof and quantitative analysis of these effects are still needed. This study employs solid-state NMR (ssNMR) to investigate how SNAC interacts with hydrated DMPC liposomes, measuring enhancements in membrane fluidity across interfacial and transmembrane regions. The methodology involves analyzing phosphate lipid headgroups and acyl chains using static 31P chemical shift anisotropy and 2H quadrupolar coupling measurements alongside 1H and 13C magic angle spinning NMR for motional averaging of 1H-1H and 1H-13C dipolar couplings. Our findings indicate an overall increase in the uniaxial motion of phospholipids with SNAC in a PE concentration-dependent manner. It boosts lipid headgroup dynamics and enhancement plateaus at 25% between 24 and 72 mM concentrations. SNAC effectively enhances the fluidity of the hydrophobic center by 43% at 72 mM PE concentration, more significantly than the interfacial region. It is worth noting that the extent of liposome dissolution and conversion to micelles increases as SNAC concentration rises. Including a model peptide drug, octreotide, introduces a competitive equilibrium in this complex PE-lipid-peptide system, further influencing membrane dynamics for peptide permeation. Interestingly, the membrane enhancement does not show the expected plateau, and a less significant lipid mobility increase is observed in the presence of octreotide, suggesting a less substantial impact compared to peptide-free systems, which is likely due to peptide-PE interactions that consume monomeric SNAC, reducing its interaction with the lipid membrane. This study provides the first quantitative and site-specific ssNMR measurements of membrane mobility influenced by one representative PE as a snapshot of PE lipid interaction in a liposome model, demonstrating how peptide drugs modulate competitive equilibria and PE-induced lipid dynamics.
Collapse
Affiliation(s)
- Jing Ling
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Ryan Schroder
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - W Peter Wuelfing
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - John Higgins
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Filippos Kesisoglou
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Allen C Templeton
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Yongchao Su
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
5
|
Zhang L, Hua Z, Fang Z, Wei J, Lin Y. Efficacy and Safety of Oral Semaglutide in the Treatment of Type 2 Diabetes: A Meta-Analysis. J Clin Pharmacol 2024; 64:1312-1325. [PMID: 38874130 DOI: 10.1002/jcph.2483] [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: 01/30/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
This study aims to systematically review the efficacy and safety of oral semaglutide in the treatment of type 2 diabetes mellitus (T2DM) and provide a basis for the rational use of the drug in clinical practice. From the database's inception until February 2023, a systematic search was conducted in PubMed, Embase, the Cochrane Library, Web of Science, China National Knowledge Infrastructure, Wanfang Database, and China Science and Technology Journal Database to identify randomized controlled trials (RCTs) comparing the efficacy of oral semaglutide at dosages of 3, 7, and 14 mg (trial group) against placebo or other positive control drugs (control group) for the treatment of T2DM. Following literature screening and data extraction, the bias risk assessment tool in the Cochrane reviewer handbook 5.1.0 was used to evaluate the literature quality. Meta-analysis was carried out with RevMan 5.4 software. A total of 10 RCTs with 9541 patients were included. The meta-analysis results revealed that compared with placebo or positive control drugs (empagliflozin, sitagliptin, liraglutide, and dulaglutide), oral semaglutide significantly reduced the hemoglobin A1c (HbA1c) in patients (compared to placebo, 3 mg [MD = -0.61%, 95% CI (-0.89, -0.34)], 7 mg [MD = -1.12%, 95% CI (-1.45, -0.79)], 14 mg [MD = -1.08%, 95% CI (-1.32, -0.85)]; compared to positive control drugs (7 mg [MD = -0.26%, 95% CI (-0.38, -0.15)], 14 mg [MD = -0.37%, 95% CI (-0.52, -0.23)]). Oral semaglutide also showed certain advantages over placebo or positive control drugs in terms of weight loss, HbA1c reduction achievement rate, fasting plasma glucose level, and body mass index with overall dose-dependent efficacy. The incidence of nausea, diarrhea, and vomiting caused by oral semaglutide was higher than that of the placebo or positive control drugs, and the incidence of appetite decrease or constipation was higher than that of the placebo. Severe or symptomatic hypoglycemic episodes were reduced compared to positive control drugs. Oral semaglutide has definite clinical benefits of reducing blood glucose, body weight, reducing the risk of hypoglycemia, and with good safety.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zixin Hua
- Department of Clinical Pharmacy, Capital Medical University, Beijing, China
| | - Zhenwei Fang
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Juanjuan Wei
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yang Lin
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Bohley M, Leroux J. Gastrointestinal Permeation Enhancers Beyond Sodium Caprate and SNAC - What is Coming Next? ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400843. [PMID: 38884149 PMCID: PMC11434117 DOI: 10.1002/advs.202400843] [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: 01/23/2024] [Revised: 03/13/2024] [Indexed: 06/18/2024]
Abstract
Oral peptide delivery is trending again. Among the possible reasons are the recent approvals of two oral peptide formulations, which represent a huge stride in the field. For the first time, gastrointestinal (GI) permeation enhancers (PEs) are leveraged to overcome the main limitation of oral peptide delivery-low permeability through the intestinal epithelium. Despite some success, the application of current PEs, such as salcaprozate sodium (SNAC), sodium caprylate (C8), and sodium caprate (C10), is generally resulting in relatively low oral bioavailabilities (BAs)-even for carefully selected therapeutics. With several hundred peptide-based drugs presently in the pipeline, there is a huge unmet need for more effective PEs. Aiming to provide useful insights for the development of novel PEs, this review summarizes the biological hurdles to oral peptide delivery with special emphasis on the epithelial barrier. It describes the concepts and action modes of PEs and mentions possible new targets. It further states the benchmark that is set by current PEs, while critically assessing and evaluating emerging PEs regarding translatability, safety, and efficacy. Additionally, examples of novel PEs under preclinical and clinical evaluation and future directions are discussed.
Collapse
Affiliation(s)
- Marilena Bohley
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Jean‐Christophe Leroux
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| |
Collapse
|
7
|
Kang C, Bernaldez M, Stamatis SD, Rose JP, Sun R. Interaction between Permeation Enhancers and Lipid Bilayers. J Phys Chem B 2024; 128:1668-1679. [PMID: 38232311 DOI: 10.1021/acs.jpcb.3c06448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Permeation enhancers (PEs) are a class of molecules that interact with the epithelial membrane and transiently increase its transcellular permeability. Although there have been few clinical trials of PE coformulated drugs, the mechanism of action of PEs remains elusive. In this paper, the interaction between two archetypes of PEs [salcaprozate sodium (SNAC) and sodium caprate (C10)] and membranes is investigated with extensive all-atom molecular dynamics simulations. The simulations show that (1) the association between the neutral PEs and membranes is favored in free energy, (2) the propensity of neutral PE aggregation is larger in aqueous solution than in lipid bilayers, (3) the equilibrium distribution of neutral PEs in membranes is fast, e.g., accessible with unbiased MD simulations, and (4) the micelle of neutral PEs formed in aqueous solution does not rupture the membranes (e.g., not forming pores or breaking up the membrane) under simulation conditions. All results combined, this study indicates that PEs insert into the membranes in an equilibrium or near equilibrium process. This study lays the foundation for future investigations of how PEs impact the free energy of permeation for small molecules.
Collapse
Affiliation(s)
- Christopher Kang
- Department of Chemistry, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| | - Mabel Bernaldez
- Department of Chemistry, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| | - Stephen D Stamatis
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - John P Rose
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Rui Sun
- Department of Chemistry, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| |
Collapse
|
8
|
Asano D, Takakusa H, Nakai D. Oral Absorption of Middle-to-Large Molecules and Its Improvement, with a Focus on New Modality Drugs. Pharmaceutics 2023; 16:47. [PMID: 38258058 PMCID: PMC10820198 DOI: 10.3390/pharmaceutics16010047] [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: 10/31/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
To meet unmet medical needs, middle-to-large molecules, including peptides and oligonucleotides, have emerged as new therapeutic modalities. Owing to their middle-to-large molecular sizes, middle-to-large molecules are not suitable for oral absorption, but there are high expectations around orally bioavailable macromolecular drugs, since oral administration is the most convenient dosing route. Therefore, extensive efforts have been made to create bioavailable middle-to-large molecules or develop absorption enhancement technology, from which some successes have recently been reported. For example, Rybelsus® tablets and Mycapssa® capsules, both of which contain absorption enhancers, were approved as oral medications for type 2 diabetes and acromegaly, respectively. The oral administration of Rybelsus and Mycapssa exposes their pharmacologically active peptides with molecular weights greater than 1000, namely, semaglutide and octreotide, respectively, into systemic circulation. Although these two medications represent major achievements in the development of orally absorbable peptide formulations, the oral bioavailability of peptides after taking Rybelsus and Mycapssa is still only around 1%. In this article, we review the approaches and recent advances of orally bioavailable middle-to-large molecules and discuss challenges for improving their oral absorption.
Collapse
Affiliation(s)
- Daigo Asano
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan; (H.T.); (D.N.)
| | | | | |
Collapse
|
9
|
Davoudi S, Raemdonck K, Braeckmans K, Ghysels A. Capric Acid and Myristic Acid Permeability Enhancers in Curved Liposome Membranes. J Chem Inf Model 2023; 63:6789-6806. [PMID: 37917127 DOI: 10.1021/acs.jcim.3c00936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Liposomes are considered as advanced drug delivery systems for cancer treatment. A generation of pH-sensitive liposomes is being developed that use fatty acids (FAs) as a trigger for drug release in tumor tissues. However, FAs are also known to enhance permeability, and it is unclear whether FAs in liposomes may cause drug leakage or premature drug release. The passive permeability of the drug through the membrane of the liposome is thus a crucial factor for timely drug delivery. To investigate how the curvature and lipid composition of liposomes affect their passive permeability, coarse-grained molecular dynamics were performed. The permeability was determined with a counting method. Flat bilayers and three liposomes with varying diameters were studied, which had varying lipid compositions of dipalmitoylphosphatidylcholine, cholesterol, and deprotonated or neutral saturated FAs. The investigated permeants were water and two other small permeants, which have different free energy profiles (solubility) across the membrane. First, for the curvature effect, our results showed that curvature increases the water permeability by reducing the membrane thickness. The permeability increase for water is about a factor of 1.7 for the most curved membranes. However, a high curvature decreases permeability for permeants with free energy profiles that are a mix of wells and barriers in the headgroup region of the membrane. Importantly, the type of experimental setup is expected to play a dominant role in the permeability value, i.e., whether permeants are escaping or entering the liposomes. Second, for the composition effect, FAs decrease both the area per lipid (APL) and the membrane thickness, resulting in permeability increases of up to 55%. Cholesterol has a similar effect on the APL but has the opposite impact on membrane thickness and permeability. Therefore, FAs and cholesterol have opposing effects on permeability, with cholesterol's effect being slightly stronger in our simulated bilayers. As all permeability values were well within a factor of 2, and with liposomes usually being larger and less curved in experimental applications, it can be concluded that the passive drug release from a pH-sensitive liposome does not seem to be significantly affected by the presence of FAs.
Collapse
Affiliation(s)
- Samaneh Davoudi
- IBiTech─BioMMedA Group, Ghent University, Corneel Heymanslaan 10, Block B-Entrance 36, 9000 Gent, Belgium
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory for General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Gent, Belgium
| | - Kevin Braeckmans
- Bio-Photonic Imaging Group, Laboratory for General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Gent, Belgium
| | - An Ghysels
- IBiTech─BioMMedA Group, Ghent University, Corneel Heymanslaan 10, Block B-Entrance 36, 9000 Gent, Belgium
| |
Collapse
|
10
|
Maher S, Geoghegan C, Brayden DJ. Safety of surfactant excipients in oral drug formulations. Adv Drug Deliv Rev 2023; 202:115086. [PMID: 37739041 DOI: 10.1016/j.addr.2023.115086] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023]
Abstract
Surfactants are a diverse group of compounds that share the capacity to adsorb at the boundary between distinct phases of matter. They are used as pharmaceutical excipients, food additives, emulsifiers in cosmetics, and as household/industrial detergents. This review outlines the interaction of surfactant-type excipients present in oral pharmaceutical dosage forms with the intestinal epithelium of the gastrointestinal (GI) tract. Many surfactants permitted for human consumption in oral products reduce intestinal epithelial cell viability in vitro and alter barrier integrity in epithelial cell monolayers, isolated GI tissue mucosae, and in animal models. This suggests a degree of mis-match for predicting safety issues in humans from such models. Recent controversial preclinical research also infers that some widely used emulsifiers used in oral products may be linked to ulcerative colitis, some metabolic disorders, and cancers. We review a wide range of surfactant excipients in oral dosage forms regarding their interactions with the GI tract. Safety data is reviewed across in vitro, ex vivo, pre-clinical animal, and human studies. The factors that may mitigate against some of the potentially abrasive effects of surfactants on GI epithelia observed in pre-clinical studies are summarised. We conclude with a perspective on the overall safety of surfactants in oral pharmaceutical dosage forms, which has relevance for delivery system development.
Collapse
Affiliation(s)
- Sam Maher
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland.
| | - Caroline Geoghegan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
11
|
Hundahl AC, Weller A, Larsen JB, Hjørringgaard CU, Hansen MB, Mündler AK, Knuhtsen A, Kristensen K, Arnspang EC, Andresen TL, Mortensen KI, Marie R. Quantitative live-cell imaging of lipidated peptide transport through an epithelial cell layer. J Control Release 2023; 355:122-134. [PMID: 36724849 DOI: 10.1016/j.jconrel.2023.01.066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/10/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023]
Abstract
Oral drug delivery increases patient compliance and is thus the preferred administration route for most drugs. However, for biologics the intestinal barrier greatly limits the absorption and reduces their bioavailability. One strategy employed to improve on this is chemical modification of the biologic through the addition of lipid side chains. While it has been established that lipidation of peptides can increase transport, a mechanistic understanding of this effect remains largely unexplored. To pursue this mechanistic understanding, end-point detection of biopharmaceuticals transported through a monolayer of fully polarized epithelial cells is typically used. However, these methods are time-consuming and tedious. Furthermore, most established methods cannot be combined easily with high-resolution live-cell fluorescence imaging that could provide a mechanistic insight into cellular uptake and transport. Here we address this challenge by developing an axial PSF deconvolution scheme to quantify the transport of peptides through a monolayer of Caco-2 cells using single-cell analysis with live-cell confocal fluorescence microscopy. We then measure the known cross-barrier transport of several compounds in our model and compare the results with results obtained in an established microfluidic model finding similar transport phenotypes. This verifies that already after two days the Caco-2 cells in our model form a tight monolayer and constitute a functional barrier model. We then apply this assay to investigate the effects of side chain lipidation of the model peptide drug salmon calcitonin (sCT) modified with 4‑carbon and 8‑carbon-long fatty acid chains. Furthermore, we compare that with experiments performed at lower temperature and using inhibitors for some endocytotic pathways to pinpoint how lipidation length modifies the main avenues for the transport. We thus show that increasing the length of the lipid chain increases the transport of the drug significantly but also makes endocytosis the primary transport mechanism in a short-term cell culture model.
Collapse
Affiliation(s)
- Adam Coln Hundahl
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Arjen Weller
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Jannik Bruun Larsen
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Claudia U Hjørringgaard
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Morten B Hansen
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Ann-Kathrin Mündler
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Astrid Knuhtsen
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Kasper Kristensen
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Eva C Arnspang
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Thomas Lars Andresen
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Kim I Mortensen
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark
| | - Rodolphe Marie
- Technical University of Denmark, Department of Health Technology, Ørsteds Plads, Building 345C, DK-2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
12
|
Kommineni N, Sainaga Jyothi VGS, Butreddy A, Raju S, Shapira T, Khan W, Angsantikul P, Domb AJ. SNAC for Enhanced Oral Bioavailability: An Updated Review. Pharm Res 2023; 40:633-650. [PMID: 36539668 DOI: 10.1007/s11095-022-03459-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
The delivery of proteins and peptides via an oral route poses numerous challenges to improve the oral bioavailability and patient compliance. To overcome these challenges, as well as to improve the permeation of proteins and peptides via intestinal mucosa, several chemicals have been studied such as surfactants, fatty acids, bile salts, pH modifiers, and chelating agents, amongst these medium chain fatty acid like C10 (sodium caprate) and Sodium N-[8-(2-hydroxybenzoyl) amino] caprylate (SNAC) and its derivatives that have been well studied from a clinical perspective. This current review enumerates the challenges involved in protein and peptide delivery via the oral route, i.e., non-invasive routes of protein and peptide administration. This review also covers the chemistry behind SNAC and toxicity as well as mechanisms to enhance the oral delivery of clinically proven molecules like simaglutide and other small molecules under clinical development, as well as other permeation enhancers for efficient delivery of proteins and peptides.
Collapse
Affiliation(s)
- Nagavendra Kommineni
- Center for Biomedical Research, Population Council, New York, NY, 10065, USA.
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India.
| | - Vaskuri G S Sainaga Jyothi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Arun Butreddy
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, Oxford, MS, 38677, USA
| | - Saka Raju
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Tovi Shapira
- School of Pharmacy and Faculty of Medicine, The Hebrew University of Jerusalem, Hadassah Medical Center, Ein Kerem Campus, 91120, Jerusalem, Israel
| | - Wahid Khan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
- Natco Research Centre, NATCO Pharma Limited, Hyderabad, 500018, India
| | - Pavimol Angsantikul
- Center for Biomedical Research, Population Council, New York, NY, 10065, USA
| | - Abraham J Domb
- School of Pharmacy and Faculty of Medicine, The Hebrew University of Jerusalem, Hadassah Medical Center, Ein Kerem Campus, 91120, Jerusalem, Israel.
| |
Collapse
|
13
|
Wibel R, Jörgensen AM, Laffleur F, Spleis H, Claus V, Bernkop-Schnürch A. Oral delivery of calcitonin-ion pairs: In vivo proof of concept for a highly lipophilic counterion. Int J Pharm 2023; 631:122476. [PMID: 36528188 DOI: 10.1016/j.ijpharm.2022.122476] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/28/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Hydrophobic ion pairing and subsequent incorporation into self-emulsifying drug delivery systems (SEDDS) is a promising strategy to orally deliver hydrophilic macromolecular drugs. Within this study, hydrophobic ion pairs (HIP) between salmon calcitonin (sCT) and highly lipophilic sulfosuccinate counterions were formed and compared to frequently applied commercially available counterions. Bis(isotridecyl) sulfosuccinate resulted in HIPs of the highest lipophilicity and in significantly higher solubility in lipophilic co-solvents. Thus, bis(isotridecyl) sulfosuccinate allowed efficient solubilization of sCT in a SEDDS preconcentrate based on a lipophilic co-solvent and an indigestible lipid, but omitting hydrophilic co-solvents. In addition to the increased solubility in the lipidic matrix, markedly reduced dissociation in biorelevant media resulted in high distribution coefficients between oil droplet and FaSSGF or FaSSIF (logD) of 2.98 ± 0.12 or 2.77 ± 0.14, respectively. The composition of the lipidic matrix preserved integrity of the oil droplets after emulsification and subsequent lipolysis, allowing to fully exploit the potential of the HIP attributed to the high logD. Oral administration of the HIP-loaded SEDDS resulted in an excellent relative pharmacological activity of 13.8 ± 5.6 % measured as hypocalcaemic effect in rats.
Collapse
Affiliation(s)
- Richard Wibel
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Arne Matteo Jörgensen
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Flavia Laffleur
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Helen Spleis
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria; Thiomatrix Forschungs-und Beratungs GmbH, Trientlgasse, 65, 6020 Innsbruck, Austria
| | - Victor Claus
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria; Thiomatrix Forschungs-und Beratungs GmbH, Trientlgasse, 65, 6020 Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria.
| |
Collapse
|
14
|
Twarog C, Fattal E, Noiray M, Illel B, Brayden D, Taverna M, Hillaireau H. Characterization of the physicochemical interactions between exenatide and two intestinal permeation enhancers: sodium caprate (C 10) and salcaprozate sodium (SNAC). Int J Pharm 2022; 626:122131. [PMID: 36028084 DOI: 10.1016/j.ijpharm.2022.122131] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/12/2022] [Accepted: 08/18/2022] [Indexed: 10/15/2022]
Abstract
A common approach to tackle the poor intestinal membrane permeability of peptides after oral administration is to formulate them with a permeation enhancer (PE). Increased oral bioavailability for oral peptide candidates has been reported from clinical trials when either salcaprozate sodium (SNAC) or sodium caprate (C10) is incorporated in the formulation. However, little is known about how they physically interact with peptides in solution. Our objective was to compare the biophysical interactions between the GLP-1 analogue exenatide (Byetta®, Lilly), and C10 or SNAC using a variety of advanced analytical techniques. First, critical micelle concentration was measured in different buffers for both PEs. Dynamic light scattering (DLS) measurements revealed specific supramolecular structures arising from exenatide-PE association. Surface plasmon resonance (SPR) indicated the formation of exenatide-PE complexes with a high contribution from non-specific interactions and rapid binding kinetics, resulting in overall low affinities. DLS and isothermal titration calorimetry (ITC) were used to examine the supramolecular organization of the PEs, and revealed thermodynamic signatures characterized by unfavourable enthalpic contributions compensated by favourable entropic ones, but with low-affinity estimates in water (KD in the 10-100 µM range). With affinity capillary electrophoresis (ACE), weak interactions between exenatide and SNAC or C10 were confirmed in saline, with a dissociation constant around 10 µM and 30 µM respectively. In biorelevant intestinal media, the bile salts in FaSSIF and FeSSIF further reduced the binding of both agents to exenatide (KD ≈ 100 µM), indicating that the interaction between the PEs and exenatide might be inhibited by bile salts in the GI lumen. This study suggests that the interactions of both PEs with exenatide follow a similar non-covalent mechanism and are of low affinity.
Collapse
Affiliation(s)
- Caroline Twarog
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France
| | - Elias Fattal
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France
| | - Magali Noiray
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France
| | - Brigitte Illel
- Drug Product Development, Sanofi Research and Development, Montpellier, France
| | - David Brayden
- UCD School of Veterinary Medicine, Belfield, Dublin 4, Ireland; UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Myriam Taverna
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France
| | - Hervé Hillaireau
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France.
| |
Collapse
|
15
|
Weng H, Hu L, Hu L, Zhou Y, Wang A, Wang N, Li W, Zhu C, Guo S, Yu M, Gan Y. The complexation of insulin with sodium N‐[8‐(2‐hydroxybenzoyl)amino]‐caprylate for enhanced oral delivery: Effects of concentration, ratio, and pH. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
16
|
Tran H, ElSayed MEH. Progress and limitations of oral peptide delivery as a potentially transformative therapy. Expert Opin Drug Deliv 2022; 19:163-178. [PMID: 35255753 DOI: 10.1080/17425247.2022.2051476] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION The oral delivery of peptides offers advantages over the injectable route of administration due to patient convenience. However, oral delivery remains challenging due to physiological barriers. Numerous formulation technologies have been developed to overcome these challenges, and understanding the advantages and limitations of each technology is important for the development of new delivery systems to enable oral delivery of peptides designed for parenteral administration. AREAS COVERED This review summarizes key learnings from the use of permeation enhancers (PEs) for oral peptide delivery associated with solid dosage form optimization to maximize the PE effect. Furthermore, we will highlight the most recent emerging delivery strategies to improve oral peptide bioavailability such as nanoparticles, self-emulsifying drug delivery systems, gut shuttles, and ingestible devices. In addition, advantages and limitations of these technologies will be compared with the permeation enhancer technology. EXPERT OPINION Despite the success of permeation enhancer technology in the FDA approved oral peptide products for gastric and intestinal delivery, oral peptide delivery is still facing the immense challenge of low-to-single digit oral bioavailability and the impact of food and water intake on oral absorption. Optimization of drug product attributes such as dissolution kinetics is critical to overcome spreading and dilution effects in vivo to improve permeation enhancer efficacy. The next frontiers to substantially increase oral bioavailability and transform injectable peptides to oral deliverables may be ingestible devices and gut shuttles. In addition, ingestible devices may have potential to overcome the impact of food on oral bioavailability. However, clinical studies are necessary to inform the safety and efficacy of these emerging technologies.
Collapse
|
17
|
Berg S, Kärrberg L, Suljovic D, Seeliger F, Söderberg M, Perez-Alcazar M, Van Zuydam N, Abrahamsson B, Hugerth AM, Davies N, Bergström CAS. Impact of Intestinal Concentration and Colloidal Structure on the Permeation-Enhancing Efficiency of Sodium Caprate in the Rat. Mol Pharm 2022; 19:200-212. [PMID: 34928160 PMCID: PMC8728734 DOI: 10.1021/acs.molpharmaceut.1c00724] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
![]()
In this work, we
set out to better understand how the permeation
enhancer sodium caprate (C10) influences the intestinal absorption
of macromolecules. FITC-dextran 4000 (FD4) was selected as a model
compound and formulated with 50–300 mM C10. Absorption was
studied after bolus instillation of liquid formulation to the duodenum
of anesthetized rats and intravenously as a reference, whereafter
plasma samples were taken and analyzed for FD4 content. It was found
that the AUC and Cmax of FD4 increased
with increasing C10 concentration. Higher C10 concentrations were
associated with an increased and extended absorption but also increased
epithelial damage. Depending on the C10 concentration, the intestinal
epithelium showed significant recovery already at 60–120 min
after administration. At the highest studied C10 concentrations (100
and 300 mM), the absorption of FD4 was not affected by the colloidal
structures of C10, with similar absorption obtained when C10 was administered
as micelles (pH 8.5) and as vesicles (pH 6.5). In contrast, the FD4
absorption was lower when C10 was administered at 50 mM formulated
as micelles as compared to vesicles. Intestinal dilution of C10 and
FD4 revealed a trend of decreasing FD4 absorption with increasing
intestinal dilution. However, the effect was smaller than that of
altering the total administered C10 dose. Absorption was similar when
the formulations were prepared in simulated intestinal fluids containing
mixed micelles of bile salts and phospholipids and in simple buffer
solution. The findings in this study suggest that in order to optimally
enhance the absorption of macromolecules, high (≥100 mM) initial
intestinal C10 concentrations are likely needed and that both the
concentration and total dose of C10 are important parameters.
Collapse
Affiliation(s)
- Staffan Berg
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, BMC P.O. Box 580, SE-751 23 Uppsala, Sweden.,Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Lillevi Kärrberg
- Animal Sciences and Technologies, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Denny Suljovic
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, BMC P.O. Box 580, SE-751 23 Uppsala, Sweden
| | - Frank Seeliger
- Cardiovascular, Renal and Metabolism Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Magnus Söderberg
- Cardiovascular, Renal and Metabolism Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Marta Perez-Alcazar
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Natalie Van Zuydam
- Data Science and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Andreas M Hugerth
- Ferring Pharmaceuticals A/S Global Pharmaceutical R&D, 2300 Copenhagen, Denmark
| | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Christel A S Bergström
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, BMC P.O. Box 580, SE-751 23 Uppsala, Sweden
| |
Collapse
|
18
|
Kneiszl R, Hossain S, Larsson P. In Silico-Based Experiments on Mechanistic Interactions between Several Intestinal Permeation Enhancers with a Lipid Bilayer Model. Mol Pharm 2022; 19:124-137. [PMID: 34913341 PMCID: PMC8728740 DOI: 10.1021/acs.molpharmaceut.1c00689] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 01/07/2023]
Abstract
Oral administration of drugs is generally considered convenient and patient-friendly. However, oral administration of biological drugs exhibits low oral bioavailability (BA) due to enzymatic degradation and low intestinal absorption. A possible approach to circumvent the low BA of oral peptide drugs is to coformulate the drugs with permeation enhancers (PEs). PEs have been studied since the 1960s and are molecules that enhance the absorption of hydrophilic molecules with low permeability over the gastrointestinal epithelium. In this study, we investigated the impact of six PEs on the structural properties of a model membrane using molecular dynamics (MD) simulations. The PEs included were the sodium salts of the medium chain fatty acids laurate, caprate, and caprylate and the caprylate derivative SNAC─all with a negative charge─and neutral caprate and neutral sucrose monolaurate. Our results indicated that the PEs, once incorporated into the membrane, could induce membrane leakiness in a concentration-dependent manner. Our simulations suggest that a PE concentration of at least 70-100 mM is needed to strongly affect transcellular permeability. The increased aggregation propensity seen for neutral PEs might provide a molecular-level mechanism for the membrane disruptions seen at higher concentrations in vivo. The ability for neutral PEs to flip-flop across the lipid bilayer is also suggestive of possible intracellular modes of action other than increasing membrane fluidity. Taken together, our results indicate that MD simulations are useful for gaining insights relevant to the design of oral dosage forms based around permeability enhancer molecules.
Collapse
Affiliation(s)
- Rosita Kneiszl
- Department
of Pharmacy, Uppsala University, Husargatan 3, Uppsala 751 23, Sweden
- The
Swedish Drug Delivery Center (SweDeliver), Uppsala University, Husargatan 3, Uppsala 751 23, Sweden
| | - Shakhawath Hossain
- Department
of Pharmacy, Uppsala University, Husargatan 3, Uppsala 751 23, Sweden
- The
Swedish Drug Delivery Center (SweDeliver), Uppsala University, Husargatan 3, Uppsala 751 23, Sweden
| | - Per Larsson
- Department
of Pharmacy, Uppsala University, Husargatan 3, Uppsala 751 23, Sweden
- The
Swedish Drug Delivery Center (SweDeliver), Uppsala University, Husargatan 3, Uppsala 751 23, Sweden
| |
Collapse
|
19
|
Chede LS, Wagner BA, Buettner GR, Donovan MD. Electron Spin Resonance Evaluation of Buccal Membrane Fluidity Alterations by Sodium Caprylate and L-Menthol. Int J Mol Sci 2021; 22:ijms221910708. [PMID: 34639049 PMCID: PMC8509842 DOI: 10.3390/ijms221910708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
The ability of sodium caprylate and l-menthol to fluidize phospholipid bilayers composed of lipids simulating the buccal epithelium was investigated using electron spin resonance (ESR) to evaluate the action of these agents as permeation enhancers. 5-Doxyl stearic acid (5-DSA) and 16-doxyl stearic acid (16-DSA) were used as spin labels to identify alterations in membrane fluidity near the polar head groups or inner acyl regions of the lipid bilayer, respectively. The molecular motion of both 5-DSA and 16-DSA showed increased disorder near the polar and inner hydrophobic regions of the bilayer in the presence of sodium caprylate suggesting fluidization in both the regions, which contributes to its permeation enhancing effects. L-menthol decreased the order parameter for 16-DSA, showing membrane fluidization only in the inner acyl regions of the bilayer, which also corresponded to its weaker permeation enhancing effects. The rapid evaluation of changes in fluidity of the bilayer in the presence of potential permeation enhancers using ESR enables improved selection of effective permeation enhancers and enhancer combinations based on their effect on membrane fluidization.
Collapse
Affiliation(s)
- Laxmi Shanthi Chede
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52242, USA;
| | - Brett A. Wagner
- Free Radical Radiation Biology, The University of Iowa, Iowa City, IA 52242, USA; (B.A.W.); (G.R.B.)
| | - Garry R. Buettner
- Free Radical Radiation Biology, The University of Iowa, Iowa City, IA 52242, USA; (B.A.W.); (G.R.B.)
| | - Maureen D. Donovan
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52242, USA;
- Correspondence:
| |
Collapse
|
20
|
Formulation strategies to improve the efficacy of intestinal permeation enhancers . Adv Drug Deliv Rev 2021; 177:113925. [PMID: 34418495 DOI: 10.1016/j.addr.2021.113925] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023]
Abstract
The use of chemical permeation enhancers (PEs) is the most widely tested approach to improve oral absorption of low permeability active agents, as represented by peptides. Several hundred PEs increase intestinal permeability in preclinical bioassays, yet few have progressed to clinical testing and, of those, only incremental increases in oral bioavailability (BA) have been observed. Still, average BA values of ~1% were sufficient for two recent FDA approvals of semaglutide and octreotide oral formulations. PEs are typically screened in static in vitro and ex-vivo models where co-presentation of active agent and PE in high concentrations allows the PE to alter barrier integrity with sufficient contact time to promote flux across the intestinal epithelium. The capacity to maintain high concentrations of co-presented agents at the epithelium is not reached by standard oral dosage forms in the upper GI tract in vivo due to dilution, interference from luminal components, fast intestinal transit, and possible absorption of the PE per se. The PE-based formulations that have been assessed in clinical trials in either immediate-release or enteric-coated solid dosage forms produce low and variable oral BA due to these uncontrollable physiological factors. For PEs to appreciably increase intestinal permeability from oral dosage forms in vivo, strategies must facilitate co-presentation of PE and active agent at the epithelium for a sustained period at the required concentrations. Focusing on peptides as examples of a macromolecule class, we review physiological impediments to optimal luminal presentation, discuss the efficacy of current PE-based oral dosage forms, and suggest strategies that might be used to improve them.
Collapse
|
21
|
Antipin IS, Alfimov MV, Arslanov VV, Burilov VA, Vatsadze SZ, Voloshin YZ, Volcho KP, Gorbatchuk VV, Gorbunova YG, Gromov SP, Dudkin SV, Zaitsev SY, Zakharova LY, Ziganshin MA, Zolotukhina AV, Kalinina MA, Karakhanov EA, Kashapov RR, Koifman OI, Konovalov AI, Korenev VS, Maksimov AL, Mamardashvili NZ, Mamardashvili GM, Martynov AG, Mustafina AR, Nugmanov RI, Ovsyannikov AS, Padnya PL, Potapov AS, Selektor SL, Sokolov MN, Solovieva SE, Stoikov II, Stuzhin PA, Suslov EV, Ushakov EN, Fedin VP, Fedorenko SV, Fedorova OA, Fedorov YV, Chvalun SN, Tsivadze AY, Shtykov SN, Shurpik DN, Shcherbina MA, Yakimova LS. Functional supramolecular systems: design and applications. RUSSIAN CHEMICAL REVIEWS 2021; 90:895-1107. [DOI: 10.1070/rcr5011] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The interest in functional supramolecular systems for the design of innovative materials and technologies, able to fundamentally change the world, is growing at a high pace. The huge array of publications that appeared in recent years in the global literature calls for systematization of the structural trends inherent in the formation of these systems revealed at different molecular platforms and practically useful properties they exhibit. The attention is concentrated on the topics related to functional supramolecular systems that are actively explored in institutes and universities of Russia in the last 10–15 years, such as the chemistry of host–guest complexes, crystal engineering, self-assembly and self-organization in solutions and at interfaces, biomimetics and molecular machines and devices.The bibliography includes 1714 references.
Collapse
|
22
|
Brayden DJ, Maher S. Transient Permeation Enhancer® (TPE®) technology for oral delivery of octreotide: a technological evaluation. Expert Opin Drug Deliv 2021; 18:1501-1512. [PMID: 34128734 DOI: 10.1080/17425247.2021.1942838] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The FDA approval of oral semaglutide for type 2 diabetes (2019) and oral octreotide for acromegaly (2020) is evidence that selected niche peptides can be administered orally if formulated with selected intestinal permeation enhancers. AREAS COVERED We evaluated the oral octreotide formulation, MYCAPSSA® (Chiasma Pharmaceuticals, Needham, MA, USA). An outline of the current standard of care in acromegaly and the benefits of oral octreotide versus depot injections is provided. We discuss the Transient Permeation Enhancer (TPE®) technology used and detail the safety and efficacy data from animal models and clinical trials. EXPERT OPINION TPE® is an oily suspension of octreotide that includes a number of excipients that can transiently alter epithelial barrier integrity by opening of intestinal epithelial tight junctions arising from transcellular perturbation. Phase I studies using 20 mg octreotide capsules yielded a relative oral bioavailability of ~0.7% and primary endpoints were achieved in two Phase III studies. The oral octreotide dose required to achieve these endpoints was over 200 times that of the 0.1 mg immediate-release subcutaneous injection, a reminder of the difficulty in achieving oral absorption of macromolecules. Many acromegaly patients will prefer a convenient twice-daily oral formulation of octreotide compared to monthly depot injections.
Collapse
Affiliation(s)
- David J Brayden
- University College Dublin (UCD) School of Veterinary Medicine, UCD, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biotechnology, UCD, Belfield, Dublin 4, Ireland.,CÚRAM, the SFI Research Centre for Medical Devices, UCD, Belfield, Dublin 4, Ireland
| | - Sam Maher
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland
| |
Collapse
|
23
|
Gleeson JP, Fein KC, Whitehead KA. Oral delivery of peptide therapeutics in infants: Challenges and opportunities. Adv Drug Deliv Rev 2021; 173:112-124. [PMID: 33774115 PMCID: PMC8178217 DOI: 10.1016/j.addr.2021.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/09/2021] [Accepted: 03/18/2021] [Indexed: 12/14/2022]
Abstract
The vast majority of drugs are not designed or developed for pediatric and infant populations. Peptide drugs, which have become increasingly relevant in the past several decades, are no exception. Unfortunately, nearly all of the 60+ approved peptide drugs are formulated for injection, a particularly unfriendly mode of administration for infants. Although three peptide drugs were recently approved for oral formulations, this major advance in peptide drug delivery is available only for adults. In this review, we consider the current challenges and opportunities for the oral formulation of peptide therapeutics, specifically for infant populations. We describe the strategies that enable oral protein delivery and the potential impact of infant physiology on those strategies. We also detail the limited but encouraging progress towards 1) adapting conventional drug development and delivery approaches to infants and 2) designing novel infant-centric formulations. Together, these efforts underscore the feasibility of oral peptide delivery in infants and provide motivation to increase attention paid to this underserved area of drug delivery and formulation.
Collapse
Affiliation(s)
- John P Gleeson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | - Katherine C Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States; Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States.
| |
Collapse
|
24
|
Mollazadeh S, Sahebkar A, Shahlaei M, Moradi S. Nano drug delivery systems: Molecular dynamic simulation. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.115823] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
25
|
McCartney F, Perinelli DR, Tiboni M, Cavanagh R, Lucarini S, Filippo Palmieri G, Casettari L, Brayden DJ. Permeability-enhancing effects of three laurate-disaccharide monoesters across isolated rat intestinal mucosae. Int J Pharm 2021; 601:120593. [PMID: 33857587 DOI: 10.1016/j.ijpharm.2021.120593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/14/2021] [Accepted: 04/07/2021] [Indexed: 10/21/2022]
Abstract
Laurate (C12)-sucrose esters are established intestinal epithelial permeation enhancers (PEs) with potential for use in oral delivery. Most studies have examined blends of ester rather than specific monoesters, with little variation on the sugar moiety. To investigate the influence of varying the sugar moiety on monoester performance, we compared three monoesters: C12-sucrose, C12-lactose, and C12-trehalose. The assays were: critical micellar concentration (CMC) in Krebs-Henseleit buffer, MTS and lactate dehydrogenase assays in Caco-2 cells, transepithelial electrical resistance (TEER) and apparent permeability coefficient (Papp) of [14C] mannitol across isolated rat intestinal mucosae, and tissue histology. For CMC, the rank order was C12-trehalose (0.21 mM) < C12-sucrose (0.34 mM) < C12-lactose (0.43 mM). Exposure to Caco-2 cells for 120 min produced TC50 values in the MTS assay from 0.1 to 0.4 mM. Each ester produced a concentration-dependent decrease in TEER across rat mucosae with 80% reduction seen with 8 mM in 5 min, but C12-trehalose was less potent. C12-sucrose and C12-lactose increased the Papp of [14C] mannitol across mucosae with similar potency and efficacy, whereas C12-trehalose was not as potent or efficacious, even though it still increased flux. In the presence of the three esters, gross intestinal histology was unaffected except at 8 mM for C12-sucrose and C12-lactose. In conclusion, the three esters enhanced permeability likely via tight junction modulation in rat intestinal tissue. C12-trehalose was not quite as efficacious, but neither did it damage tissue to the same extent. All three can be considered as potential PEs to be included in oral formulations.
Collapse
Affiliation(s)
- Fiona McCartney
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Diego R Perinelli
- School of Pharmacy, University of Camerino, Via Gentile III da Varano, 62032 Camerino, MC, Italy
| | - Mattia Tiboni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento, 6, 61029 Urbino, PU, Italy
| | - Robert Cavanagh
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Simone Lucarini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento, 6, 61029 Urbino, PU, Italy
| | | | - Luca Casettari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento, 6, 61029 Urbino, PU, Italy.
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
26
|
Brayden DJ, Stuettgen V. Sodium glycodeoxycholate and sodium deoxycholate as epithelial permeation enhancers: in vitro and ex vivo intestinal and buccal bioassays. Eur J Pharm Sci 2021; 159:105737. [PMID: 33524502 DOI: 10.1016/j.ejps.2021.105737] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022]
Abstract
Bile salts were first tested as epithelial permeation enhancers (PEs) for the intestine and buccal routes over 20 years ago. They are not as popular as other PEs due to their non-specific mechanism of action and perceived toxicity potential. We revisited two of them by comparing efficacy and toxicity of sodium glycodeoxycholate (SGC) and sodium deoxycholate (DC) for both routes using in vitro and ex vivo methods. Cytotoxicity assays in Caco-2 cells revealed that both agents altered cellular parameters at concentrations >2 mM over 60 min. Both agents reduced the transepithelial resistance (TEER) and doubled the Papp of [3H]-octreotide across isolated rat colonic mucosae mounted in Ussing chambers at 10 mM concentrations. In some studies, 10 mM GDC also increased the Papp of the paracellular marker, FITC-dextran 4000 (FD4) and the fluorescent peptide, FITC-LKP, across colonic mucosae. Tissue histology was intact despite some mild perturbation at 10 mM. In the buccal epithelial cell line, TR146, changes in cell parameters were also seen at 1.5 mM over 60 min for both agents, with slightly more sensitivity seen for DC. In isolated porcine buccal epithelial mucosae, GDC was slightly more potent and efficacious than DC at increasing the Papp of [14C]-mannitol. It also increased the Papp of [3H]-octreotide and FITC-LKP by ∼3-fold across porcine buccal tissue without causing damage. Overall, GDC and DC were efficacious in intestinal and buccal models. Both cause mild perturbation leading to an increase in paracellular fluxes for hydrophilic molecules including peptides. Their moderate efficacy, low potency, and low toxicity in these models are similar to that of more established PEs in clinical trials.
Collapse
Affiliation(s)
- David J Brayden
- UCD School of Veterinary Medicine; UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Vivien Stuettgen
- UCD School of Veterinary Medicine; UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
27
|
A nanoemulsion/micelles mixed nanosystem for the oral administration of hydrophobically modified insulin. Drug Deliv Transl Res 2021; 11:524-545. [PMID: 33575972 PMCID: PMC7987602 DOI: 10.1007/s13346-021-00920-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2021] [Indexed: 01/07/2023]
Abstract
The potential of nanoemulsions for the oral administration of peptides is still in its early stage. The aim of the present work was to rationally design, develop, and fully characterize a new nanoemulsion (NE) intended for the oral administration of hydrophobically modified insulin (HM-insulin). Specific components of the NE were selected based on their enhancing permeation properties as well as their ability to improve insulin association efficiency (Miglyol 812, sodium taurocholate), stability in the intestinal fluids, and mucodiffusion (PEGylated phospholipids and poloxamer 407). The results showed that the NE co-existed with a population of micelles, forming a mixed system that exhibited a 100% of HM-insulin association efficiency. The nanosystem showed good stability and miscibility in different bio-relevant media and displayed an acceptable mucodiffusive behavior in porcine mucus. In addition, it exhibited a high interaction with cell mono-cultures (Caco -2 and C2BBe1 human colon carcinoma Caco-2 clone cells) and co-cultures (C2BBe1 human colon carcinoma Caco-2 clone/HT29-MTX cells). The internalization in Caco-2 monolayers was also confirmed by confocal microscopy. Finally, the promising in vitro behavior of the nanosystem in terms of overcoming the biological barriers of the intestinal tract was translated into a moderate, although significant, hypoglycemic response (≈ 20–30%), following intestinal administration to both healthy and diabetic rat models. Overall, this information underlines the crucial steps to address when designing peptide-based nanoformulations to successfully overcome the intestinal barriers associated to the oral modality of administration. ![]()
Collapse
|
28
|
Hossain S, Joyce P, Parrow A, Jõemetsa S, Höök F, Larsson P, Bergström CAS. Influence of Bile Composition on Membrane Incorporation of Transient Permeability Enhancers. Mol Pharm 2020; 17:4226-4240. [PMID: 32960068 PMCID: PMC7610231 DOI: 10.1021/acs.molpharmaceut.0c00668] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
![]()
Transient
permeability enhancers (PEs), such as caprylate, caprate,
and salcaprozate sodium (SNAC), improve the bioavailability of poorly
permeable macromolecular drugs. However, the effects are variable
across individuals and classes of macromolecular drugs and biologics.
Here, we examined the influence of bile compositions on the ability
of membrane incorporation of three transient PEs—caprylate,
caprate, and SNAC—using coarse-grained molecular dynamics (CG-MD).
The availability of free PE monomers, which are important near the
absorption site, to become incorporated into the membrane was higher
in fasted-state fluids than that in fed-state fluids. The simulations
also showed that transmembrane perturbation, i.e.,
insertion of PEs into the membrane, is a key mechanism by which caprylate
and caprate increase permeability. In contrast, SNAC was mainly adsorbed
onto the membrane surface, indicating a different mode of action.
Membrane incorporation of caprylate and caprate was also influenced
by bile composition, with more incorporation into fasted- than fed-state
fluids. The simulations of transient PE interaction with membranes
were further evaluated using two experimental techniques: the quartz
crystal microbalance with dissipation technique and total internal
reflection fluorescence microscopy. The experimental results were
in good agreement with the computational simulations. Finally, the
kinetics of membrane insertion was studied with CG-MD. Variation in
micelle composition affected the insertion rates of caprate monomer
insertion and expulsion from the micelle surface. In conclusion, this
study suggests that the bile composition and the luminal composition
of the intestinal fluid are important factors contributing to the
interindividual variability in the absorption of macromolecular drugs
administered with transient PEs.
Collapse
Affiliation(s)
- Shakhawath Hossain
- Department of Pharmacy, Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden.,The Swedish Drug Delivery Forum (SDDF), Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden
| | - Paul Joyce
- Division of Biological Physics, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Albin Parrow
- Department of Pharmacy, Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden
| | - Silver Jõemetsa
- Division of Biological Physics, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Fredrik Höök
- Division of Biological Physics, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Per Larsson
- Department of Pharmacy, Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden.,The Swedish Drug Delivery Forum (SDDF), Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden
| | - Christel A S Bergström
- Department of Pharmacy, Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden.,The Swedish Drug Delivery Forum (SDDF), Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden
| |
Collapse
|
29
|
Maher S, Geoghegan C, Brayden DJ. Intestinal permeation enhancers to improve oral bioavailability of macromolecules: reasons for low efficacy in humans. Expert Opin Drug Deliv 2020; 18:273-300. [PMID: 32937089 DOI: 10.1080/17425247.2021.1825375] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Intestinal permeation enhancers (PEs) are substances that transiently alter the intestinal epithelial barrier to facilitate permeation of macromolecules with low oral bioavailability (BA). While a number of PEs have progressed to clinical testing in conventional formulations with macromolecules, there has been only low single digit increases in oral BA, irrespective of whether the drug met primary or secondary clinical endpoints. AREAS COVERED This article considers the causes of sub-optimal BA of macromolecules from PE dosage forms and suggests approaches that may improve performance in humans. EXPERT OPINION Permeation enhancement is most effective when the PE is co-localized with the macromolecule at the epithelial surface. Conditions in the GI tract impede optimal co-localization. Novel delivery systems that limit dilution and spreading of the PE and macromolecule in the small intestine have attempted to replicate promising enhancement efficacy observed in static drug delivery models.
Collapse
Affiliation(s)
- Sam Maher
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Caroline Geoghegan
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
30
|
Dumont C, Beloqui A, Miolane C, Bourgeois S, Préat V, Fessi H, Jannin V. Solid lipid nanocarriers diffuse effectively through mucus and enter intestinal cells - but where is my peptide? Int J Pharm 2020; 586:119581. [PMID: 32603838 DOI: 10.1016/j.ijpharm.2020.119581] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 01/06/2023]
Abstract
Peptides are therapeutic molecules with high potential to treat a wide variety of diseases. They are large hydrophilic compounds for which absorption is limited by the intestinal epithelial border covered by mucus. This study aimed to evaluate the potential of Hydrophobic Ion Pairing combined with Solid Lipid Nanoparticles (SLN) and Nanostructured Lipid Carriers (NLC) to improve peptide transport across the intestinal border using Caco-2 cell monolayers (enterocyte-like model) and Caco-2/HT29-MTX co-cultured monolayers (mucin-secreting model). A Hydrophobic Ion Pair (HIP) was formed between Leuprolide (LEU), a model peptide, and sodium docusate. The marked increase in peptide lipophilicity enabled high encapsulation efficiencies in both NLC (84%) and SLN (85%). After co-incubation with the nanoparticles, confocal microscopy images of the cell monolayers demonstrated particles internalization and ability to cross mucus. Flow cytometry measurements confirmed that 82% of incubated SLN and 99% of NLC were internalized by Caco-2 cells. However, LEU transport across cell monolayers was not improved by the nanocarriers. Indeed, combination of particles platelet-shape and HIP low stability in the transport medium led to LEU burst release in this environment. Improvement of peptide lipidization should maintain encapsulation and enable benefit from nanocarriers enhanced intestinal transport.
Collapse
Affiliation(s)
- Camille Dumont
- Gattefossé SAS, 36 Chemin de Genas, 69804 Saint-Priest Cedex, France; Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69100 Villeurbanne, France
| | - Ana Beloqui
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Cédric Miolane
- Gattefossé SAS, 36 Chemin de Genas, 69804 Saint-Priest Cedex, France
| | - Sandrine Bourgeois
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69100 Villeurbanne, France; Univ Lyon, Université Claude Bernard, Lyon 1, ISPB-Faculté de Pharmacie de Lyon, F-69008 Lyon, France
| | - Véronique Préat
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Hatem Fessi
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69100 Villeurbanne, France; Univ Lyon, Université Claude Bernard, Lyon 1, ISPB-Faculté de Pharmacie de Lyon, F-69008 Lyon, France
| | - Vincent Jannin
- Gattefossé SAS, 36 Chemin de Genas, 69804 Saint-Priest Cedex, France.
| |
Collapse
|
31
|
Brayden DJ. Per Artursson's Major Contributions to the Caco-2 Cell Literature in Pharmaceutical Sciences. J Pharm Sci 2020; 110:12-16. [PMID: 32860800 DOI: 10.1016/j.xphs.2020.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/20/2020] [Accepted: 08/20/2020] [Indexed: 01/18/2023]
Abstract
This edition of the Journal of Pharmaceutical Sciences is dedicated to the wonderful career of Per Artursson from the Uppsala University. My Commentary focusses on Per's major contributions to the Caco-2 cell literature over the past 30 years. Two especially influential papers have been cited more than 1000 times out of a total citation count of almost 30,000 and a h-index of 93 (Google Scholar), making Per one of the most cited and influential Pharmaceutical scientists of his generation. The Caco-2 field to which Per contributed so many advances has informed the community on key areas including predictive drug fluxes across the intestine, metabolism by intestinal epithelia, the role of transporters during flux, enantiomer-selective flux, excipient interaction with tight junctions, and nanoparticle uptake by enterocytes. In this pioneering work, Per has been careful to emphasise that Caco-2 monolayers have limitations and are a model of the human small intestine where observations must be backed up with in vitro tissue and in vivo work. Throughout, he has paid great attention to detail in methodology, as reflected by co-authorship of two Nature Protocols on Caco-2 assays. The article briefly assesses some of the most important milestones in Per's published Caco-2 research.
Collapse
Affiliation(s)
- David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
32
|
Brayden D, Hill T, Fairlie D, Maher S, Mrsny R. Systemic delivery of peptides by the oral route: Formulation and medicinal chemistry approaches. Adv Drug Deliv Rev 2020; 157:2-36. [PMID: 32479930 DOI: 10.1016/j.addr.2020.05.007] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
In its 33 years, ADDR has published regularly on the po5tential of oral delivery of biologics especially peptides and proteins. In the intervening period, analysis of the preclinical and clinical trial failures of many purported platform technologies has led to reflection on the true status of the field and reigning in of expectations. Oral formulations of semaglutide, octreotide, and salmon calcitonin have completed Phase III trials, with oral semaglutide being approved by the FDA in 2019. The progress made with oral peptide formulations based on traditional permeation enhancers is against a background of low and variable oral bioavailability values of ~1%, leading to a current perception that only potent peptides with a viable cost of synthesis can be realistically considered. Desirable features of candidates should include a large therapeutic index, some stability in the GI tract, a long elimination half-life, and a relatively low clearance rate. Administration in nanoparticle formats have largely disappointed, with few prototypes reaching clinical trials: insufficient particle loading, lack of controlled release, low epithelial particle uptake, and lack of scalable synthesis being the main reasons for discontinuation. Disruptive technologies based on engineered devices promise improvements, but scale-up and toxicology aspects are issues to address. In parallel, medicinal chemists are synthesizing stable hydrophobic macrocyclic candidate peptides of lower molecular weight and with potential for greater oral bioavailability than linear peptides, but perhaps without the same requirement for elaborate drug delivery systems. In summary, while there have been advances in understanding the limitations of peptides for oral delivery, low membrane permeability, metabolism, and high clearance rates continue to hamper progress.
Collapse
|
33
|
Twarog C, Liu K, O'Brien PJ, Dawson KA, Fattal E, Illel B, Brayden DJ. A head-to-head Caco-2 assay comparison of the mechanisms of action of the intestinal permeation enhancers: SNAC and sodium caprate (C 10). Eur J Pharm Biopharm 2020; 152:95-107. [PMID: 32387703 DOI: 10.1016/j.ejpb.2020.04.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/15/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
Salcaprozate sodium (SNAC) and sodium caprate (C10) are the two leading intestinal permeation enhancers (PEs) in oral peptide formulations in clinical trials. There is debate over their mechanism of action on intestinal epithelia. The aims were: (i) to compare their effects on the barrier function by measuring transepithelial electrical resistance (TEER), permeability of FITC-4000 (FD4) across Caco-2 monolayers, and on immunohistochemistry of tight junction (TJ)-associated proteins; and (ii) to compare cellular parameters using conventional end-point cytotoxicity assays and quantitative high content analysis (HCA) of multiple sub-lethal parameters in Caco-2 cells. C10 (8.5 mM) reversibly reduced TEER and increased FD4 permeability across monolayers, whereas SNAC had no effects on either parameter except at cytotoxic concentrations. C10 exposure induced reorganization of three TJ proteins, whereas SNAC only affected claudin-5 localization. High concentrations of C10 and SNAC were required to cause end-point toxicology changes in vitro. SNAC was less potent than C10 at inducing lysosomal and nuclear changes and plasma membrane perturbation. In parallel, HCA revealed that both agents displayed detergent-like features that reflect initial membrane fluidization followed by changes in intracellular parameters. In conclusion, FD4 permeability increases in monolayers in response to C10 were in the range of concentrations that altered end-point cytotoxicity and HCA parameters. For SNAC, while HCA parameters were also altered in a similar overall pattern as C10, they did not lead to increased paracellular flux. These assays show that both agents are primarily surfactants, but C10 has additional TJ-opening effects. While these in vitro assays illucidate their epithelial mechanism of action, clinical experience suggests that they over-estimate their toxicology in the dynamic intestinal environment.
Collapse
Affiliation(s)
- Caroline Twarog
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kai Liu
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland; UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Peter J O'Brien
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kenneth A Dawson
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland; UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Elias Fattal
- School of Pharmacy, Institut Galien, CNRS, Univ. Paris-Sud, Univ. Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Brigitte Illel
- Drug Product Development, Small Molecules Oral Platform, Sanofi Research and Development, Montpellier, France
| | - David J Brayden
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
34
|
Stuettgen V, Brayden DJ. Investigations of Piperazine Derivatives as Intestinal Permeation Enhancers in Isolated Rat Intestinal Tissue Mucosae. AAPS JOURNAL 2020; 22:33. [PMID: 31989362 DOI: 10.1208/s12248-020-0416-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/24/2019] [Indexed: 12/31/2022]
Abstract
A limiting factor for oral delivery of macromolecules is low intestinal epithelial permeability. 1-Phenylpiperazine (PPZ), 1-(4-methylphenyl) piperazine (1-4-MPPZ) and 1-methyl-4-phenylpiperazine (1-M-4-PPZ) have emerged as potential permeation enhancers (PEs) from a screen carried out by others in Caco-2 monolayers. Here, their efficacy, mechanism of action and potential for epithelial toxicity were further examined in Caco-2 cells and isolated rat intestinal mucosae. Using high-content analysis, PPZ and 1-4-MPPZ decreased mitochondrial membrane potential and increased plasma membrane potential in Caco-2 cells to a greater extent than 1-M-4-PPZ. The Papp of the paracellular marker, [14C]-mannitol, and of the peptide, [3H]-octreotide, was measured across rat colonic mucosae following apical addition of the three piperazines. PPZ and 1-4-MPPZ induced a concentration-dependent decrease in transepithelial electrical resistance (TEER) and an increase in the Papp of [14C]-mannitol without causing histological damage. 1-M-4-PPZ was without effect. The piperazines caused the Krebs-Henseleit buffer pH to become alkaline, which partially attenuated the increase in Papp of [14C]-mannitol caused by PPZ and 1-4-MPPZ. Only addition of 1-4-MPPZ increased the Papp of [3H]-octreotide. Pre-incubation of mucosae with two 5-HT4 receptor antagonists, a loop diuretic and a myosin light chain kinase inhibitor, reduced the permeation enhancement capacity of PPZ and 1-4-MPP for [14C]-mannitol. 1-4-MPPZ holds most promise as a PE, but intestinal physiology may also be impacted due to multiple mechanisms of action.
Collapse
Affiliation(s)
- V Stuettgen
- School of Veterinary Medicine and Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - D J Brayden
- School of Veterinary Medicine and Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
35
|
Jurišić Dukovski B, Juretić M, Bračko D, Randjelović D, Savić S, Crespo Moral M, Diebold Y, Filipović-Grčić J, Pepić I, Lovrić J. Functional ibuprofen-loaded cationic nanoemulsion: Development and optimization for dry eye disease treatment. Int J Pharm 2019; 576:118979. [PMID: 31870964 DOI: 10.1016/j.ijpharm.2019.118979] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/31/2022]
Abstract
Inflammation plays a key role in dry eye disease (DED) affecting millions of people worldwide. Non-steroidal anti-inflammatory drugs (NSAIDs) can be used topically to act on the inflammatory component of DED, but their limited aqueous solubility raises formulation issues. The aim of this study was development and optimization of functional cationic nanoemulsions (NEs) for DED treatment, as a formulation approach to circumvent solubility problems, prolong drug residence at the ocular surface and stabilize the tear film. Ibuprofen was employed as the model NSAID, chitosan as the cationic agent, and lecithin as the anionic surfactant enabling chitosan incorporation. Moreover, lecithin is a mixture of phospholipids including phosphatidylcholine and phosphatidylethanolamine, two constituents of the natural tear film important for its stability. NEs were characterized in terms of droplet size, polydispersity index, zeta-potential, pH, viscosity, osmolarity, surface tension, entrapment efficiency, stability, sterilizability and in vitro release. NEs mucoadhesive properties were tested rheologically after mixing with mucin dispersion. Biocompatibility was assessed employing 3D HCE-T cell-based model and ex vivo model using porcine corneas. The results of our study pointed out the NE formulation with 0.05% (w/w) chitosan as the lead formulation with physicochemical properties adequate for ophthalmic application, mucoadhesive character and excellent biocompatibility.
Collapse
Affiliation(s)
- Bisera Jurišić Dukovski
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Pharmaceutical Technology, Zagreb, Croatia
| | - Marina Juretić
- R&D, PLIVA Croatia Ltd, TEVA Group Member, Zagreb, Croatia
| | - Danka Bračko
- R&D, PLIVA Croatia Ltd, TEVA Group Member, Zagreb, Croatia
| | - Danijela Randjelović
- University of Belgrade, Institute of Chemistry, Technology and Metallurgy, Department of Microelectronic Technologies, Belgrade, Serbia
| | - Snežana Savić
- University of Belgrade, Faculty of Pharmacy, Department of Pharmaceutical Technology and Cosmetology, Belgrade, Serbia
| | - Mario Crespo Moral
- University of Valladolid, Institute of Applied Ophthalmo-Biology (IOBA), Valladolid, Spain
| | - Yolanda Diebold
- University of Valladolid, Institute of Applied Ophthalmo-Biology (IOBA), Valladolid, Spain; Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain
| | - Jelena Filipović-Grčić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Pharmaceutical Technology, Zagreb, Croatia
| | - Ivan Pepić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Pharmaceutical Technology, Zagreb, Croatia
| | - Jasmina Lovrić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Pharmaceutical Technology, Zagreb, Croatia.
| |
Collapse
|
36
|
Han X, Zhang E, Shi Y, Song B, Du H, Cao Z. Biomaterial-tight junction interaction and potential impacts. J Mater Chem B 2019; 7:6310-6320. [PMID: 31364678 PMCID: PMC6812605 DOI: 10.1039/c9tb01081e] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The active pharmaceutical ingredients (APIs) have to cross the natural barriers and get into the blood to impart the pharmacological effects. The tight junctions (TJs) between the epithelial cells serve as the major selectively permeable barriers and control the paracellular transport of the majority of hydrophilic drugs, in particular, peptides and proteins. TJs perfectly balance the targeted transport and the exclusion of other unexpected pathogens under the normal conditions. Many biomaterials have shown the capability to open the TJs and improve the oral bioavailability and targeting efficacy of the APIs. Nevertheless, there is limited understanding of the biomaterial-TJ interactions. The opening of the TJs further poses the risk of autoimmune diseases and infections. This review article summarizes the most updated literature and presents insights into the TJ structure, the biomaterial-TJ interaction mechanism, the benefits and drawbacks of TJ disruption, and methods for evaluating such interactions.
Collapse
Affiliation(s)
- Xiangfei Han
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, USA.
| | - Ershuai Zhang
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, USA.
| | - Yuanjie Shi
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, USA.
| | - Boyi Song
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, USA.
| | - Hong Du
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, USA.
| | - Zhiqiang Cao
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, USA.
| |
Collapse
|
37
|
McCartney F, Rosa M, Brayden DJ. Evaluation of Sucrose Laurate as an Intestinal Permeation Enhancer for Macromolecules: Ex Vivo and In Vivo Studies. Pharmaceutics 2019; 11:E565. [PMID: 31683652 PMCID: PMC6921008 DOI: 10.3390/pharmaceutics11110565] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023] Open
Abstract
Oral delivery of macromolecules requires permeation enhancers (PEs) adaptable to formulation. Sucrose laurate (SL) (D1216), a food grade surfactant, was assessed in Caco-2 monolayers, isolated rat intestinal tissue mucosae, and rat intestinal instillations. Accordingly, 1 mM SL increased the apparent permeability coefficient (Papp) of [14C]-mannitol and reduced transepithelial electrical resistance (TEER) across monolayers. It altered expression of the tight junction protein, ZO-1, increased plasma membrane potential, and decreased mitochondrial membrane potential in Caco-2 cells. The concentrations that increased flux were of the same order as those that induced cytotoxicity. In rat colonic tissue mucosae, the same patterns emerged in respect to the concentration-dependent increases in paracellular marker fluxes and TEER reductions with 5 mM being the key concentration. While the histology revealed some perturbation, ion transport capacity was retained. In rat jejunal and colonic instillations, 50 and 100 mM SL co-administered with insulin induced blood glucose reductions and achieved relative bioavailability values of 2.4% and 8.9%, respectively, on a par with the gold standard PE, sodium caprate (C10). The histology of the intestinal loops revealed little damage. In conclusion, SL is a candidate PE with high potential for emulsion-based systems. The primary action is plasma membrane perturbation, leading to tight junction openings and a predominant paracellular flux.
Collapse
Affiliation(s)
- Fiona McCartney
- UCD School of Veterinary Medicine and UCD Conway Institute, University, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Mónica Rosa
- Sublimity Therapeutics, DCU Alpha Innovation Campus, Dublin, Dublin 11, Ireland.
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
38
|
Nowak E, Livney YD, Niu Z, Singh H. Delivery of bioactives in food for optimal efficacy: What inspirations and insights can be gained from pharmaceutics? Trends Food Sci Technol 2019. [DOI: 10.1016/j.tifs.2019.07.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Labrasol® is an efficacious intestinal permeation enhancer across rat intestine: Ex vivo and in vivo rat studies. J Control Release 2019; 310:115-126. [PMID: 31401199 DOI: 10.1016/j.jconrel.2019.08.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 01/04/2023]
Abstract
Labrasol® ALF (Labrasol®), is a non-ionic surfactant excipient primarily used as a solubilising agent. It was investigated here as an intestinal permeation enhancer in isolated rat colonic mucosae in Ussing chamber and in rat in situ intestinal instillations. Labrasol® comprises mono-, di- and triglycerides and mono- and di- fatty acid esters of polyethylene glycol (PEG)-8 and free PEG-8, with caprylic (C8)- and capric acid (C10) as the main fatty acids. Source components of Labrasol® as well as Labrasol® modified with either C8 or C10 as the sole fatty acid components were also tested to determine which element of Labrasol® was responsible for its permeability-enhancing properties. Labrasol® (4, 8 mg/mL) enhanced the transport of the paracellular markers, [14C] mannitol, FITC-dextran 4000, and FITC-insulin across colonic mucosae. The enhancement was non-damaging, transient, and molecular weight-dependent. The PEG ester fraction of Labrasol® also had enhancing properties. When insulin was administered with Labrasol® in instillations, it had a relative bioavailability of 7% in jejunum and 12% in colon. C8- and C10 versions of Labrasol® and the PEG ester fraction also induced similar bioavailability values in jejunal instillations: 6, 5 and 7% respectively. Inhibition of lipases in instillations did not reduce the efficacy of Labrasol®, suggesting that its mechanism as a PE is not simply due to liberated medium chain fatty acids. Labrasol® acts as an efficacious intestinal permeation enhancer and has potential for use in oral formulations of macromolecules and BCS Class III molecules.
Collapse
|
40
|
Twarog C, Fattah S, Heade J, Maher S, Fattal E, Brayden DJ. Intestinal Permeation Enhancers for Oral Delivery of Macromolecules: A Comparison between Salcaprozate Sodium (SNAC) and Sodium Caprate (C 10). Pharmaceutics 2019; 11:E78. [PMID: 30781867 PMCID: PMC6410172 DOI: 10.3390/pharmaceutics11020078] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 12/31/2022] Open
Abstract
Salcaprozate sodium (SNAC) and sodium caprate (C10) are two of the most advanced intestinal permeation enhancers (PEs) that have been tested in clinical trials for oral delivery of macromolecules. Their effects on intestinal epithelia were studied for over 30 years, yet there is still debate over their mechanisms of action. C10 acts via openings of epithelial tight junctions and/or membrane perturbation, while for decades SNAC was thought to increase passive transcellular permeation across small intestinal epithelia based on increased lipophilicity arising from non-covalent macromolecule complexation. More recently, an additional mechanism for SNAC associated with a pH-elevating, monomer-inducing, and pepsin-inhibiting effect in the stomach for oral delivery of semaglutide was advocated. Comparing the two surfactants, we found equivocal evidence for discrete mechanisms at the level of epithelial interactions in the small intestine, especially at the high doses used in vivo. Evidence that one agent is more efficacious compared to the other is not convincing, with tablets containing these PEs inducing single-digit highly variable increases in oral bioavailability of payloads in human trials, although this may be adequate for potent macromolecules. Regarding safety, SNAC has generally regarded as safe (GRAS) status and is Food and Drug Administration (FDA)-approved as a medical food (Eligen®-Vitamin B12, Emisphere, Roseland, NJ, USA), whereas C10 has a long history of use in man, and has food additive status. Evidence for co-absorption of microorganisms in the presence of either SNAC or C10 has not emerged from clinical trials to date, and long-term effects from repeat dosing beyond six months have yet to be assessed. Since there are no obvious scientific reasons to prefer SNAC over C10 in orally delivering a poorly permeable macromolecule, then formulation, manufacturing, and commercial considerations are the key drivers in decision-making.
Collapse
Affiliation(s)
- Caroline Twarog
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Sarinj Fattah
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Joanne Heade
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Sam Maher
- School of Pharmacy, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland.
| | - Elias Fattal
- School of Pharmacy, Institut Galien, CNRS, Univ. Paris-Sud, Univ. Paris-Saclay, 92290 Châtenay-Malabry, France.
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
41
|
Application of Permeation Enhancers in Oral Delivery of Macromolecules: An Update. Pharmaceutics 2019; 11:pharmaceutics11010041. [PMID: 30669434 PMCID: PMC6359609 DOI: 10.3390/pharmaceutics11010041] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/29/2022] Open
Abstract
The application of permeation enhancers (PEs) to improve transport of poorly absorbed active pharmaceutical ingredients across the intestinal epithelium is a widely tested approach. Several hundred compounds have been shown to alter the epithelial barrier, and although the research emphasis has broadened to encompass a role for nanoparticle approaches, PEs represent a key constituent of conventional oral formulations that have progressed to clinical testing. In this review, we highlight promising PEs in early development, summarize the current state of the art, and highlight challenges to the translation of PE-based delivery systems into safe and effective oral dosage forms for patients.
Collapse
|
42
|
Hossain MS, Berg S, Bergström CAS, Larsson P. Aggregation Behavior of Medium Chain Fatty Acids Studied by Coarse-Grained Molecular Dynamics Simulation. AAPS PharmSciTech 2019; 20:61. [PMID: 30627943 PMCID: PMC6373435 DOI: 10.1208/s12249-018-1289-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/19/2018] [Indexed: 11/30/2022] Open
Abstract
Medium chain fatty acids (MCFA) are digestion products of lipid-rich food and lipid-based formulations, and they are used as transient permeability enhancers in formulation of poorly permeable compounds. These molecules may promote drug absorption by several different processes, including solubilization, increased membrane fluidity, and increased paracellular transport through opening of the tight junctions. Therefore, understanding the aggregation behavior of MCFAs is important. A number of studies have measured the critical micelle concentration (CMC) of MCFAs experimentally. However, CMC is highly dependent on system conditions like pH, temperature, and the ionic strength of the buffer used in different experimental techniques. In this study, we investigated the aggregation behavior of four different MCFAs using the coarse-grained molecular dynamics (CG-MD) simulations with the purpose to explore if CG-MD can be used to study MCFA interactions occurring in water. The ratio of deprotonated and non-charged MCFA molecules were manipulated to assess aggregation behavior under different pH conditions and within the box sizes of 22 × 22 × 44 nm3 and 44 nm3 for 1 μs. CMCs were calculated by performing CG-MD simulations with an increasing number of MCFAs. The resulting aggregate size distribution and number of free MCFA molecules were used to determine the CMC. The CMCs from simulations for C8, C10, and C12 were 1.8–3.5-fold lower than the respective CMCs determined experimentally by the Wilhelmy method. However, the variation of MCFA aggregate sizes and morphologies at different pH conditions is consistent with previous experimental observation. Overall, this study suggests that CG-MD is suitable for studying colloidal systems including various MCFAs.
Collapse
|
43
|
Han JR, Gu LP, Zhang RJ, Shang WH, Yan JN, McClements DJ, Wu HT, Zhu BW, Xiao H. Bioaccessibility and cellular uptake of β-carotene in emulsion-based delivery systems using scallop (Patinopecten yessoensis) gonad protein isolates: effects of carrier oil. Food Funct 2019; 10:49-60. [DOI: 10.1039/c8fo01390j] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Emulsion-based delivery systems were structured using scallop gonad protein isolates as novel food-grade emulsifiers.
Collapse
Affiliation(s)
- Jia-Run Han
- School of Food Science and Technology
- Dalian Polytechnic University
- Dalian Liaoning 116034
- China
| | - Lu-Ping Gu
- Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Jiangnan University
- Wuxi 214122
- China
| | - Ruo-Jie Zhang
- Department of Food Science
- University of Massachusetts
- Amherst
- USA
| | - Wen-Hui Shang
- School of Food Science and Technology
- Dalian Polytechnic University
- Dalian Liaoning 116034
- China
| | - Jia-Nan Yan
- School of Food Science and Technology
- Dalian Polytechnic University
- Dalian Liaoning 116034
- China
| | | | - Hai-Tao Wu
- School of Food Science and Technology
- Dalian Polytechnic University
- Dalian Liaoning 116034
- China
- National Engineering Research Center of Seafood
| | - Bei-Wei Zhu
- School of Food Science and Technology
- Dalian Polytechnic University
- Dalian Liaoning 116034
- China
- National Engineering Research Center of Seafood
| | - Hang Xiao
- Department of Food Science
- University of Massachusetts
- Amherst
- USA
| |
Collapse
|
44
|
Knudsen LB, Lau J. The Discovery and Development of Liraglutide and Semaglutide. Front Endocrinol (Lausanne) 2019; 10:155. [PMID: 31031702 PMCID: PMC6474072 DOI: 10.3389/fendo.2019.00155] [Citation(s) in RCA: 490] [Impact Index Per Article: 81.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/21/2019] [Indexed: 12/12/2022] Open
Abstract
The discovery of glucagon-like peptide-1 (GLP-1), an incretin hormone with important effects on glycemic control and body weight regulation, led to efforts to extend its half-life and make it therapeutically effective in people with type 2 diabetes (T2D). The development of short- and then long-acting GLP-1 receptor agonists (GLP-1RAs) followed. Our article charts the discovery and development of the long-acting GLP-1 analogs liraglutide and, subsequently, semaglutide. We examine the chemistry employed in designing liraglutide and semaglutide, the human and non-human studies used to investigate their cellular targets and pharmacological effects, and ongoing investigations into new applications and formulations of these drugs. Reversible binding to albumin was used for the systemic protraction of liraglutide and semaglutide, with optimal fatty acid and linker combinations identified to maximize albumin binding while maintaining GLP-1 receptor (GLP-1R) potency. GLP-1RAs mediate their effects via this receptor, which is expressed in the pancreas, gastrointestinal tract, heart, lungs, kidneys, and brain. GLP-1Rs in the pancreas and brain have been shown to account for the respective improvements in glycemic control and body weight that are evident with liraglutide and semaglutide. Both liraglutide and semaglutide also positively affect cardiovascular (CV) outcomes in individuals with T2D, although the precise mechanism is still being explored. Significant weight loss, through an effect to reduce energy intake, led to the approval of liraglutide (3.0 mg) for the treatment of obesity, an indication currently under investigation with semaglutide. Other ongoing investigations with semaglutide include the treatment of non-alcoholic fatty liver disease (NASH) and its use in an oral formulation for the treatment of T2D. In summary, rational design has led to the development of two long-acting GLP-1 analogs, liraglutide and semaglutide, that have made a vast contribution to the management of T2D in terms of improvements in glycemic control, body weight, blood pressure, lipids, beta-cell function, and CV outcomes. Furthermore, the development of an oral formulation for semaglutide may provide individuals with additional benefits in relation to treatment adherence. In addition to T2D, liraglutide is used in the treatment of obesity, while semaglutide is currently under investigation for use in obesity and NASH.
Collapse
Affiliation(s)
- Lotte Bjerre Knudsen
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
- *Correspondence: Lotte Bjerre Knudsen
| | - Jesper Lau
- Global Research Technology, Novo Nordisk A/S, Måløv, Denmark
| |
Collapse
|
45
|
Buckley ST, Bækdal TA, Vegge A, Maarbjerg SJ, Pyke C, Ahnfelt-Rønne J, Madsen KG, Schéele SG, Alanentalo T, Kirk RK, Pedersen BL, Skyggebjerg RB, Benie AJ, Strauss HM, Wahlund PO, Bjerregaard S, Farkas E, Fekete C, Søndergaard FL, Borregaard J, Hartoft-Nielsen ML, Knudsen LB. Transcellular stomach absorption of a derivatized glucagon-like peptide-1 receptor agonist. Sci Transl Med 2018; 10:10/467/eaar7047. [DOI: 10.1126/scitranslmed.aar7047] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 10/22/2018] [Indexed: 11/02/2022]
Abstract
Oral administration of therapeutic peptides is hindered by poor absorption across the gastrointestinal barrier and extensive degradation by proteolytic enzymes. Here, we investigated the absorption of orally delivered semaglutide, a glucagon-like peptide-1 analog, coformulated with the absorption enhancer sodiumN-[8-(2-hydroxybenzoyl) aminocaprylate] (SNAC) in a tablet. In contrast to intestinal absorption usually seen with small molecules, clinical and preclinical dog studies revealed that absorption of semaglutide takes place in the stomach, is confined to an area in close proximity to the tablet surface, and requires coformulation with SNAC. SNAC protects against enzymatic degradation via local buffering actions and only transiently enhances absorption. The mechanism of absorption is shown to be compound specific, transcellular, and without any evidence of effect on tight junctions. These data have implications for understanding how highly efficacious and specific therapeutic peptides could be transformed from injectable to tablet-based oral therapies.
Collapse
|
46
|
Application of Box-Behnken experimental design for the formulation and optimisation of selenomethionine-loaded chitosan nanoparticles coated with zein for oral delivery. Int J Pharm 2018; 551:257-269. [DOI: 10.1016/j.ijpharm.2018.08.050] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/23/2018] [Accepted: 08/24/2018] [Indexed: 02/07/2023]
|
47
|
Garcia J, Fernández-Blanco Á, Teixidó M, Sánchez-Navarro M, Giralt E. d-Polyarginine Lipopeptides as Intestinal Permeation Enhancers. ChemMedChem 2018; 13:2045-2052. [DOI: 10.1002/cmdc.201800428] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/30/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Josep Garcia
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Baldiri Reixac 10 08028 Barcelona Spain
| | - Álvaro Fernández-Blanco
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Baldiri Reixac 10 08028 Barcelona Spain
- Cellular & Systems Neurobiology, Systems Biology Program, Centre for Genomic Regulation (CRG); The Barcelona Institute of Science and Technology; Dr. Aiguader 88 08003 Barcelona Spain
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Baldiri Reixac 10 08028 Barcelona Spain
| | - Macarena Sánchez-Navarro
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Baldiri Reixac 10 08028 Barcelona Spain
| | - Ernest Giralt
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Baldiri Reixac 10 08028 Barcelona Spain
- University of Barcelona; Department of Inorganic and Organic Chemistry; Spain
| |
Collapse
|
48
|
Birch D, Diedrichsen RG, Christophersen PC, Mu H, Nielsen HM. Evaluation of drug permeation under fed state conditions using mucus-covered Caco-2 cell epithelium. Eur J Pharm Sci 2018. [DOI: 10.1016/j.ejps.2018.02.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
49
|
Gleeson JP, Frías JM, Ryan SM, Brayden DJ. Sodium caprate enables the blood pressure-lowering effect of Ile-Pro-Pro and Leu-Lys-Pro in spontaneously hypertensive rats by indirectly overcoming PepT1 inhibition. Eur J Pharm Biopharm 2018; 128:179-187. [PMID: 29684535 DOI: 10.1016/j.ejpb.2018.04.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 03/06/2018] [Accepted: 04/19/2018] [Indexed: 01/02/2023]
Abstract
The tripeptides, Ile-Pro-Pro (IPP) and Leu-Lys-Pro (LKP), inhibit angiotensin-converting enzyme (ACE) resulting in lowered blood pressure. Our hypothesis was that the medium chain fatty acid permeation enhancer, sodium caprate (C10), may prevent the decrease in permeability of the tripeptides when PepT1 is inhibited by glycyl-sarcosine (Gly-Sar), a situation that may occur in the presence of food hydrolysates. Using Caco-2 monolayers and isolated rat jejunal tissue, the apparent permeability coefficients (Papp) of [3H]-IPP and [3H]-LKP were assessed in the presence of Gly-Sar with and without C10. Gly-Sar decreased the Papp of both tripeptides across monolayers and isolated jejunal tissue, but C10 restored it. C10 likely increased the paracellular permeability of the tripeptides, as indicated by immunofluorescence changes in tight junction proteins in Caco-2 monolayers accompanied by a concentration-dependent decrease in transepithelial electrical resistance (TEER). [3H]-IPP and [3H]-LKP were orally-gavaged to normal rats with Gly-Sar, C10, or with a mixture. Plasma levels of both peptides were reduced by Gly-Sar to less than half that of the levels detected in its absence, but were restored when C10 was co-administered. In spontaneously hypertensive rats (SHRs), unlabelled IPP and LKP lowered blood pressure when delivered either by i.v. or oral routes. Oral gavage of Gly-Sar reduced the hypotensive action of peptides in SHRs, but the effect was restored in the presence of C10. In conclusion, there was a reduction in the hypotensive effects of IPP and LKP in SHRs when intestinal PepT1 was inhibited by Gly-Sar, but C10 may circumvent this by enhancing paracellular permeability.
Collapse
Affiliation(s)
- John P Gleeson
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA(1)
| | - Jesús M Frías
- Environmental Science and Health Institute, Dublin Institute of Technology, Grangegorman, Dublin 7, Ireland
| | - Sinéad M Ryan
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
50
|
Ghadiri M, Canney F, Pacciana C, Colombo G, Young PM, Traini D. The use of fatty acids as absorption enhancer for pulmonary drug delivery. Int J Pharm 2018; 541:93-100. [DOI: 10.1016/j.ijpharm.2018.02.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 01/15/2018] [Accepted: 02/15/2018] [Indexed: 01/12/2023]
|