1
|
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
|
2
|
Surowiec RK, Saldivar R, Rai RK, Metzger CE, Jacobson AM, Allen MR, Wallace JM. Ex vivo exposure to calcitonin or raloxifene improves mechanical properties of diseased bone through non-cell mediated mechanisms. Bone 2023; 173:116805. [PMID: 37196853 PMCID: PMC10330631 DOI: 10.1016/j.bone.2023.116805] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/01/2023] [Accepted: 05/12/2023] [Indexed: 05/19/2023]
Abstract
Raloxifene (RAL) reduces clinical fracture risk despite modest effects on bone mass and density. This reduction in fracture risk may be due to improved material level-mechanical properties through a non-cell mediated increase in bone hydration. Synthetic salmon calcitonin (CAL) has also demonstrated efficacy in reducing fracture risk with only modest bone mass and density improvements. This study aimed to determine if CAL could modify healthy and diseased bone through cell-independent mechanisms that alter hydration similar to RAL. 26-week-old male C57BL/6 mice induced with chronic kidney disease (CKD) beginning at 16 weeks of age via 0.2 % adenine-laced casein-based (0.9 % P, 0.6 % C) chow, and their non-CKD control littermates (Con), were utilized. Upon sacrifice, right femora were randomly assigned to the following ex vivo experimental groups: RAL (2 μM, n = 10 CKD, n = 10 Con), CAL (100 nM, n = 10 CKD, n = 10 Con), or Vehicle (VEH; n = 9 CKD, n = 9 Con). Bones were incubated in PBS + drug solution at 37 °C for 14 days using an established ex vivo soaking methodology. Cortical geometry (μCT) was used to confirm a CKD bone phenotype, including porosity and cortical thinning, at sacrifice. Femora were assessed for mechanical properties (3-point bending) and bone hydration (via solid state nuclear magnetic resonance spectroscopy with magic angle spinning (ssNMR)). Data were analyzed by two-tailed t-tests (μCT) or 2-way ANOVA for main effects of disease, treatment, and their interaction. Tukey's post hoc analyses followed a significant main effect of treatment to determine the source of the effect. Imaging confirmed a cortical phenotype reflective of CKD, including lower cortical thickness (p < 0.0001) and increased cortical porosity (p = 0.02) compared to Con. In addition, CKD resulted in weaker, less deformable bones. In CKD bones, ex vivo exposure to RAL or CAL improved total work (+120 % and +107 %, respectively; p < 0.05), post-yield work (+143 % and +133 %), total displacement (+197 % and +229 %), total strain (+225 % and +243 %), and toughness (+158 % and +119 %) vs. CKD VEH soaked bones. Ex vivo exposure to RAL or CAL did not impact any mechanical properties in Con bone. Matrix-bound water by ssNMR showed CAL treated bones had significantly higher bound water compared to VEH treated bones in both CKD and Con cohorts (p = 0.001 and p = 0.01, respectively). RAL positively modulated bound water in CKD bone compared to VEH (p = 0.002) but not in Con bone. There were no significant differences between bones soaked with CAL vs. RAL for any outcomes measured. RAL and CAL improve important post-yield properties and toughness in a non-cell mediated manner in CKD bone but not in Con bones. While RAL treated CKD bones had higher matrix-bound water content in line with previous reports, both Con and CKD bones exposed to CAL had higher matrix-bound water. Therapeutic modulation of water, specifically the bound water fraction, represents a novel approach to improving mechanical properties and potentially reducing fracture risk.
Collapse
Affiliation(s)
- Rachel K Surowiec
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA; Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Rosario Saldivar
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Epidemiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ratan K Rai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Corinne E Metzger
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Andrea M Jacobson
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.
| | - Matthew R Allen
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
| | - Joseph M Wallace
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
3
|
Gastrointestinal Permeation Enhancers for the Development of Oral Peptide Pharmaceuticals. Pharmaceuticals (Basel) 2022; 15:ph15121585. [PMID: 36559036 PMCID: PMC9781085 DOI: 10.3390/ph15121585] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Recently, two oral-administered peptide pharmaceuticals, semaglutide and octreotide, have been developed and are considered as a breakthrough in peptide and protein drug delivery system development. In 2019, the Food and Drug Administration (FDA) approved an oral dosage form of semaglutide developed by Novo Nordisk (Rybelsus®) for the treatment of type 2 diabetes. Subsequently, the octreotide capsule (Mycapssa®), developed through Chiasma's Transient Permeation Enhancer (TPE) technology, also received FDA approval in 2020 for the treatment of acromegaly. These two oral peptide products have been a significant success; however, a major obstacle to their oral delivery remains the poor permeability of peptides through the intestinal epithelium. Therefore, gastrointestinal permeation enhancers are of great relevance for the development of subsequent oral peptide products. Sodium salcaprozate (SNAC) and sodium caprylate (C8) have been used as gastrointestinal permeation enhancers for semaglutide and octreotide, respectively. Herein, we briefly review two approved products, Rybelsus® and Mycapssa®, and discuss the permeation properties of SNAC and medium chain fatty acids, sodium caprate (C10) and C8, focusing on Eligen technology using SNAC, TPE technology using C8, and gastrointestinal permeation enhancement technology (GIPET) using C10.
Collapse
|
4
|
Tran H, Patel PJ, Aburub A, Sperry A, Estwick S, ElSayed MEH, -Mannan AD. Identification of a Multi-Component Formulation for Intestinal Delivery of a GLP-1/Glucagon Co-agonist Peptide. Pharm Res 2022; 39:2555-2567. [PMID: 36050547 DOI: 10.1007/s11095-022-03372-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/14/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE Oral delivery of therapeutic peptides has been challenging due to multiple physiological factors and physicochemical properties of peptides. We report a systematic approach to identify formulation compositions combining a permeation enhancer and a peptidase inhibitor that minimize proteolytic degradation and increase absorption of a peptide across the small intestine. METHODS An acylated glucagon-like peptide-1/glucagon co-agonist peptide (4.5 kDa) was selected as a model peptide. Proteolytic stability of the peptide was investigated in rat and pig SIF. Effective PEs and multiple component formulations were identified in rats. Relative bioavailability of the peptide was determined in minipigs via intraduodenal administration (ID) of enteric capsules. RESULTS The peptide degraded rapidly in the rat and pig SIF. Citric acid, SBTI, and SBTCI inhibited the enzymatic degradation. The peptide self-associated into trimers in solution, however, addition of PEs monomerized the peptide. C10 was the most effective PE among tested PEs (DPC, LC, rhamnolipid, C12-maltosides, and SNAC) to improve intestinal absorption of the peptide in the rat IJ-closed loop model. A combination of C10 and SBTI or SBTCI increased the peptide exposure 5-tenfold compared to the exposure with the PE alone in the rat IJ-cannulated model, and achieved 1.06 ± 0.76% bioavailability in minipigs relative to subcutaneous via ID administration using enteric capsules. CONCLUSION We identified SBTI and C10 as an effective peptidase inhibitor and PE for intestinal absorption of the peptide. The combination of SBTI and C10 addressed the peptide physiochemical properties and provides a formulation strategy to achieve intestinal delivery of this peptide.
Collapse
Affiliation(s)
- Huyen Tran
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA.
| | - Phenil J Patel
- Synthetic Molecule Design and Development, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Aktham Aburub
- Synthetic Molecule Design and Development, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Andrea Sperry
- Department of Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Selina Estwick
- Department of Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Mohamed E H ElSayed
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Amita Datta -Mannan
- Exploratory Medicine and Pharmacology, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA.
| |
Collapse
|
5
|
Chen H, Lu Y, Shi S, Zhang Q, Cao X, Sun L, An D, Zhang X, Kong X, Liu J. Design and Development of a New Glucagon-Like Peptide-1 Receptor Agonist to Obtain High Oral Bioavailability. Pharm Res 2022; 39:1891-1906. [PMID: 35698011 DOI: 10.1007/s11095-022-03265-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/18/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Semaglutide is the only oral GLP-1 RA in the market, but oral bioavailability is generally limited in range of 0.4-1%. In this study, a new GLP-1RA named SHR-2042 was developed to gain higher oral bioavailability than semaglutide. METHOD Self-association of SHR-2042, semaglutide and liraglutide were assessed using SEC-MALS. The intestinal perfusion test in SD rats was used to select permeation enhancers (PEs) including SNAC, C10 and LCC. ITC, CD and DLS were used to explore the interaction between SHR-2042 and SNAC. Gastric administrated test in SD rats was used to screen SHR-2042 granules with different SHR-2042/SNAC ratios. The oral bioavailability of SHR-2042 was studied in rats and monkeys. RESULT The designed GLP-1RA, SHR-2042, gives a better solubility and lipophilicity than semaglutide. While it forms a similar oligomer with that of semaglutide. During the selection of PEs, SNAC shows better exposure than the other competing PEs including C10 and LCC. SHR-2042 and SNAC bind quickly and exhibit hydrophobic interaction. SNAC could promote monomerization of SHR-2042 and form micelles to trap the monomerized SHR-2042. The oral bioavailability of SHR-2042 paired with SNAC is 0.041% (1:0, w/w), 0.083% (1:10, w/w), 0.32% (1:30, w/w) and 2.83% (1:60, w/w) in rats. And the oral bioavailability of SHR-2042 matched with SNAC is 3.39% (1:30, w/w) in monkeys, which is over 10 times higher than that of semaglutide. CONCLUSION We believe that the design and development of oral SHR-2042 will provide a new way to design more and more GLP-1RAs with high oral bioavailability in the future.
Collapse
Affiliation(s)
- Hao Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Yun Lu
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Shuai Shi
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Qiang Zhang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Xiaoli Cao
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Lei Sun
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Dong An
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Xiaojie Zhang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Xianglin Kong
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Jianping Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
6
|
|
7
|
Kaneb A, Berardino K, Hanukaai JS, Rooney K, Kaye AD. Calcitonin (FORTICAL, MIACALCIN) for the treatment of vertebral compression fractures. Orthop Rev (Pavia) 2021; 13:24976. [PMID: 34745472 DOI: 10.52965/001c.24976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 12/28/2022] Open
Abstract
Purpose of Review Osteoporosis is a common condition affecting the musculoskeletal system. It carries with it increased risks of fracture in many areas of the body, leading to reduced quality of life, limited mobility, and other long-term implications such as chronic pain. Vertebral compression fractures are a common development in patients with osteoporosis. Current treatment options focus on reducing pain; preventative methods are somewhat limited and focus on minimizing risk factors for the development of osteoporosis. In this review, we explore the use of calcitonin (FORTICAL, MIACALCIN) to treat vertebral compression fractures (VCFs). Recent Findings Osteoporosis had a prevalence of more than 10% in the United States in 2010. The CDC estimates that nearly 25% of women over age 65 have findings of osteoporosis, which include low spinal bone mass. The condition is highly prevalent and, in an aging U.S. population, quite clinically relevant. Risk factors for development include advanced age, cigarette smoking, medications, reduced physical activity, and low calcium and vitamin D intake. Family history may also play a role. Diagnosis is made based on bone mineral density.Standard therapy for VCFs in osteoporosis includes analgesic medications, such as NSAIDs and biphosphonates, and surgical intervention. NSAIDs address the chronic pain that is a common long-term effect of VCFs. Biphosphonates have recently been used to attempt to halt the progression and provide prevention. Surgical interventions such as balloon kyphoplasty and vertebroplasty are typically reserved for patients who have failed other methods.Calcitonin is a peptide naturally produced by the human body, released from the parathyroid gland. It binds to osteoclasts, inhibiting them from inducing bone resorption. By relatively unknown mechanisms, it also appears to cause endorphin release and mitigate pain. Clinical data has shown safety and efficacy for exogenous calcitonin in reducing bone turnover and reducing VCF-induced pain. Summary Osteoporosis is a common condition that can lead to complications such as vertebral compression fractures. It can significantly impact the quality of life in many elderly Americans. There is currently no singular treatment, but calcitonin has recently been explored as a possible option for minimizing pain and reducing disease progression. Further studies are needed to understand its preventative benefits fully.
Collapse
Affiliation(s)
- Alicia Kaneb
- Georgetown University School of Medicine, Washington D.C
| | | | | | - Kelsey Rooney
- Louisiana State University Health Science Center, Shreveport, LA
| | | |
Collapse
|
8
|
Ahmadipour S, Varshosaz J, Hashemibeni B, Safaeian L, Manshaei M, Sarmadi A. Calcitonin-loaded octamaleimic acid-silsesquioxane nanoparticles in hydrogel scaffold support osteoinductivity in bone regeneration. Pharm Dev Technol 2020; 26:220-232. [PMID: 33258707 DOI: 10.1080/10837450.2020.1858318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Novel osteoinductive scaffolds fabricated using the benefits of tissue engineering techniques accompanied by utilizing drugs can accelerate bone regeneration. The purpose of this study was to load salmon calcitonin (sCT) in octamaleimic acid-silsesquioxane (OMA-POSS) nanoparticles and enrich the hydrogel scaffold based on hydroxyapatite, Gelrite® and platelet-rich plasma (PRP) for use in bone tissue engineering. The loading efficiency, release percentage, particle size and zeta potential of the nanoparticles were evaluated. The proliferation of seeded MG-63 osteoblast cells on the designed scaffold, its cytotoxicity and osteo-conductivity were studied by alkaline phosphatase measurement and Alizarin red staining. The expression of cellular osteogenic markers such as collagen 1 (COL1A1), osteocalcin (BGLAP) and osteopontin (SPP1) was examined using reverse transcription polymerase chain reaction. The results revealed that the particle size of the nanoparticles varied between 94.2 and 199.2 nm and their negative surface charge increased after drug conjugation. The osteoblast cell proliferation and calcium granule production in the optimum formulation were significantly higher in comparison with the control group (p < 0.05). Osteogenic markers increased significantly after a specific number of days of cell culture compared to the control group (p < 0.05). The results also showed the potential of the designed scaffold in bone tissue engineering.
Collapse
Affiliation(s)
- Saeedeh Ahmadipour
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Pharmaceutics, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Batool Hashemibeni
- Department of Anatomical Sciences, Faculty of Medicine, Torabinejad Dental Research Center, Dental School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Safaeian
- Department of Pharmacology and Toxicology, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maziar Manshaei
- Dental Research Center, Dental Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Akram Sarmadi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
9
|
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
|
10
|
Sadeghi S, Lee WK, Kong SN, Shetty A, Drum CL. Oral administration of protein nanoparticles: An emerging route to disease treatment. Pharmacol Res 2020; 158:104685. [DOI: 10.1016/j.phrs.2020.104685] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 01/20/2023]
|
11
|
Sun L, Le Z, He S, Liu J, Liu L, Leong KW, Mao HQ, Liu Z, Chen Y. Flash Fabrication of Orally Targeted Nanocomplexes for Improved Transport of Salmon Calcitonin across the Intestine. Mol Pharm 2020; 17:757-768. [PMID: 32011888 DOI: 10.1021/acs.molpharmaceut.9b00827] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Salmon calcitonin (sCT) is a potent calcium-regulating peptide hormone and widely applied for the treatment of some bone diseases clinically. However, the therapeutic usefulness of sCT is hindered by the frequent injection required, owing to its short plasma half-life and therapeutic need for a high dose. Oral delivery is a popular modality of administration for patients because of its convenience to self-administration and high patient compliance, while orally administered sCT remains a great challenge currently due to the existence of multiple barriers in the gastrointestinal (GI) tract. Here, we introduced an orally targeted delivery system to increase the transport of sCT across the intestine through both the paracellular permeation route and the bile acid pathway. In this system, sCT-based glycol chitosan-taurocholic acid conjugate (GC-T)/dextran sulfate (DS) ternary nanocomplexes (NC-T) were produced by a flash nanocomplexation (FNC) process in a kinetically controlled mode. The optimized NC-T exhibited well-controlled properties with a uniform and sub-60 nm hydrodynamic diameter, high batch-to-batch reproducibility, good physical or chemical stability, as well as sustained drug release behaviors. The studies revealed that NC-T could effectively improve the intestinal uptake and permeability, owing to its surface functionalization with the taurocholic acid ligand. In the rat model, orally administered NC-T showed an obvious hypocalcemia effect and a relative oral bioavailability of 10.9%. An in vivo assay also demonstrated that NC-T induced no observable side effect after long-term oral administration. As a result, the orally targeted nanocomplex might be a promising candidate for improving the oral transport of therapeutic peptides.
Collapse
Affiliation(s)
- Lilong Sun
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology, Sun Yat-sen University, Guangzhou 510275, China.,Department of Biomedical Engineering, School of Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhicheng Le
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Shuran He
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Jingyan Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Lixin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Hai-Quan Mao
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Department of Biomedical Engineering and Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Zhijia Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
12
|
Wang X, Wang Y, Yan M, Liang X, Zhao N, Ma Y, Gao Y. Thermosensitive Hydrogel Based on Poly(2-Ethyl-2-Oxazoline)-Poly(D,L-Lactide)-Poly(2-Ethyl-2-Oxazoline) for Sustained Salmon Calcitonin Delivery. AAPS PharmSciTech 2020; 21:71. [PMID: 31953574 DOI: 10.1208/s12249-020-1619-1] [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: 10/13/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022] Open
Abstract
This study developed a thermosensitive hydrogel based on poly(2-ethyl-2-oxazoline)-poly(D,L-lactide)-poly(2-ethyl-2-oxazoline) (PPP) for the delivery of salmon calcitonin to improve the hypocalcemic effect. The tube inversion and rheological tests revealed that the copolymer solution underwent temperature-dependent sol-gel-sol transitions. Observation by scanning electron microscopy (SEM) showed that the hydrogel exhibited a porous three-dimensional network. The swelling test demonstrated that there was a maximum swelling ratio at low temperature (25°C) as compared with the high temperature (37°C). In vitro release revealed that the PPP hydrogel were capable of sustained release of salmon calcitonin (sCT). The in vivo biodegradability study indicated the good degradability of PPP hydrogel. More importantly, the in vivo retention time of the hydrogel in situ was significantly prolonged after subcutaneous injection of the PPP hydrogel compared to the F127 hydrogel. In vivo pharmacodynamics analysis showed that the hypocalcemic effect of both PPP and F127 hydrogel was significantly greater than that of sCT solution, and the mean serum Ca reduction effect could be maintained for 24 h of PPP hydrogel, indicating that PPP hydrogel could achieve a significant enhanced hypocalcemic effect. In conclusion, the PPP hydrogel has been shown to be prospective as a controlled release carrier for injection delivery of protein drugs.
Collapse
|
13
|
Preparation and characterization of solid lipid nanoparticles loaded with salmon calcitonin phospholipid complex. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.05.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
14
|
Talegaonkar S, Bhattacharyya A. Potential of Lipid Nanoparticles (SLNs and NLCs) in Enhancing Oral Bioavailability of Drugs with Poor Intestinal Permeability. AAPS PharmSciTech 2019; 20:121. [PMID: 30805893 DOI: 10.1208/s12249-019-1337-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 02/04/2019] [Indexed: 11/30/2022] Open
Abstract
Lipid-based drug delivery systems has become a popular choice for oral delivery of lipophilic drugs with dissolution rate limited oral absorption. Lipids are known to enhance oral bioavailability of poorly water-soluble drugs in multiple ways like facilitating dissolution as micellar solution, enhancing the lymphatic uptake and acting as inhibitors of efflux transporters. Lipid nanoparticles are matrix type lipid-based carrier systems which can effectively encapsulate both lipophilic and hydrophilic drugs. Lipid nanoparticles namely solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) are versatile drug delivery system and can be used for multiple routes of delivery like parenteral, topical, ocular, transdermal, and oral. Lipid nanoparticles are particularly attractive vehicles for peroral delivery of drugs with oral bioavailability problems as they are composed of lipid excipients which are cheap, easily available, and non-toxic; manufacturing technique is simple and readily scalable for large-scale production; the formulations provide controlled release of active components and have no stability issue. A large number of drugs have been incorporated into lipid nanoparticles with the objective of overcoming their poor oral bioavailability. This review tries to assess the potential of lipid nanoparticles for enhancing the oral bioavailability of drugs with permeability limited oral absorption such as drugs belonging to class IV of Biopharmaceutic Classification System (BCS) and protein and peptide drugs and also discusses the mechanism behind the bioavailability enhancement and safety issues related to such delivery systems.
Collapse
|
15
|
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
|
16
|
Shang H, Chen X, Liu Y, Yu L, Li J, Ding J. Cucurbit[7]-assisted sustained release of human calcitonin from thermosensitive block copolymer hydrogel. Int J Pharm 2017; 527:52-60. [DOI: 10.1016/j.ijpharm.2017.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/18/2017] [Accepted: 05/03/2017] [Indexed: 11/28/2022]
|
17
|
Xing C, Xing JF, Ge ZQ. Inhibition of proteases activity in intestine needs a sustainable acidic environment rather than a transient. Drug Dev Ind Pharm 2017; 43:1648-1655. [PMID: 28541760 DOI: 10.1080/03639045.2017.1328433] [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: 10/19/2022]
Abstract
α-Chymotrypsin (α-CT) and trypsin are important components of the enzymatic barrier. They could degrade the therapeutic proteins and peptides, inhibit their activity consequently, and thereby reduce their oral bioavailability. Acidic agents, as one type of indirect protease inhibitors, have shown proof of concept in clinical trials. We report here the inactivated proteases due to acid influence can be reactivated immediately by environmental pH recovery regardless of how long the inactivation last. To keep the inactivation time of proteases for 4-5 h, we designed and prepared a sustained-release tablet containing citric acid (CA) which can effectively reduce the pH below 5.0 and maintain it for 5 h in the dissolution-reaction medium. The activity of α-CT and trypsin was quantified by analyzing the residual amount of their respective substrates BTEE and TAME. More than 80% of the substrates were survived in 5.0 h of incubation, whereas the common tablet inhibited the proteases activity for only two hours in the same experimental medium. It indicates that the sustained-release tablet loaded with CA can efficiently inhibit the α-CT and trypsin activity longer than the common tablet. The results will be beneficial for designing and formulating the peroral administration of peptide and protein drugs.
Collapse
Affiliation(s)
- Chang Xing
- a Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology , Tianjin University, Education Ministry Key Laboratory of Systems Bioengineering , Tianjin , PR China
| | - Jin-Feng Xing
- a Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology , Tianjin University, Education Ministry Key Laboratory of Systems Bioengineering , Tianjin , PR China
| | - Zhi-Qiang Ge
- a Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology , Tianjin University, Education Ministry Key Laboratory of Systems Bioengineering , Tianjin , PR China
| |
Collapse
|
18
|
Lin CH, Chen CH, Lin ZC, Fang JY. Recent advances in oral delivery of drugs and bioactive natural products using solid lipid nanoparticles as the carriers. J Food Drug Anal 2017; 25:219-234. [PMID: 28911663 PMCID: PMC9332520 DOI: 10.1016/j.jfda.2017.02.001] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 02/21/2017] [Indexed: 11/30/2022] Open
Abstract
Chemical and enzymatic barriers in the gastrointestinal (GI) tract hamper the oral delivery of many labile drugs. The GI epithelium also contributes to poor permeability for numerous drugs. Drugs with poor aqueous solubility have difficulty dissolving in the GI tract, resulting in low bioavailability. Nanomedicine provides an opportunity to improve the delivery efficiency of orally administered drugs. Solid lipid nanoparticles (SLNs) are categorized as a new generation of lipid nanoparticles consisting of a complete solid lipid matrix. SLNs used for oral administration offer several benefits over conventional formulations, including increased solubility, enhanced stability, improved epithelium permeability and bioavailability, prolonged half-life, tissue targeting, and minimal side effects. The nontoxic excipients and sophisticated material engineering of SLNs tailor the controllable physicochemical properties of the nanoparticles for GI penetration via mucosal or lymphatic transport. In this review, we highlight the recent progress in the development of SLNs for disease treatment. Recent application of oral SLNs includes therapies for cancers, central nervous system-related disorders, cardiovascular-related diseases, infection, diabetes, and osteoporosis. In addition to drugs that may be active cargos in SLNs, some natural compounds with pharmacological activity are also suitable for SLN encapsulation to enhance oral bioavailability. In this article, we systematically introduce the concepts and amelioration mechanisms of the nanomedical techniques for drug- and natural compound-loaded SLNs.
Collapse
|
19
|
Maher S, Mrsny RJ, Brayden DJ. Intestinal permeation enhancers for oral peptide delivery. Adv Drug Deliv Rev 2016; 106:277-319. [PMID: 27320643 DOI: 10.1016/j.addr.2016.06.005] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 12/15/2022]
Abstract
Intestinal permeation enhancers (PEs) are one of the most widely tested strategies to improve oral delivery of therapeutic peptides. This article assesses the intestinal permeation enhancement action of over 250 PEs that have been tested in intestinal delivery models. In depth analysis of pre-clinical data is presented for PEs as components of proprietary delivery systems that have progressed to clinical trials. Given the importance of co-presentation of sufficiently high concentrations of PE and peptide at the small intestinal epithelium, there is an emphasis on studies where PEs have been formulated with poorly permeable molecules in solid dosage forms and lipoidal dispersions.
Collapse
|
20
|
Karamanidou T, Bourganis V, Kammona O, Kiparissides C. Lipid-based nanocarriers for the oral administration of biopharmaceutics. Nanomedicine (Lond) 2016; 11:3009-3032. [DOI: 10.2217/nnm-2016-0265] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Biopharmaceutics have been recognized as the drugs of choice for the treatment of several diseases, mainly due to their high selectivity and potent action. Nonetheless, their oral administration is a rather challenging problem, since their bioavailability is significantly hindered by various physiological barriers along the GI tract, including their acid-induced hydrolysis in the stomach, their enzymatic degradation throughout the GI tract and their poor mucosa permeability. Lipid-based nanocarriers represent a viable means for enhancing the oral bioavailability of biomolecules while diminishing toxicity-related issues. The present review describes the main physiological barriers limiting the oral bioavailability of macromolecules and highlights recent advances in the field of lipid-based carriers as well as the respective lipid intestinal absorption mechanisms.
Collapse
Affiliation(s)
- Theodora Karamanidou
- Department of Chemical Engineering, Aristotle University of Thessaloniki, PO Box 472, 54124 Thessaloniki, Greece
| | - Vassilis Bourganis
- Department of Chemical Engineering, Aristotle University of Thessaloniki, PO Box 472, 54124 Thessaloniki, Greece
| | - Olga Kammona
- Chemical Process & Energy Resources Institute, Centre for Research & Technology Hellas, PO Box 60361, 57001 Thessaloniki, Greece
| | - Costas Kiparissides
- Department of Chemical Engineering, Aristotle University of Thessaloniki, PO Box 472, 54124 Thessaloniki, Greece
- Chemical Process & Energy Resources Institute, Centre for Research & Technology Hellas, PO Box 60361, 57001 Thessaloniki, Greece
| |
Collapse
|
21
|
Cole LE, Vargo-Gogola T, Roeder RK. Targeted delivery to bone and mineral deposits using bisphosphonate ligands. Adv Drug Deliv Rev 2016; 99:12-27. [PMID: 26482186 DOI: 10.1016/j.addr.2015.10.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/01/2015] [Accepted: 10/09/2015] [Indexed: 01/07/2023]
Abstract
The high concentration of mineral present in bone and pathological calcifications is unique compared with all other tissues and thus provides opportunity for targeted delivery of pharmaceutical drugs, including radiosensitizers and imaging probes. Targeted delivery enables accumulation of a high local dose of a therapeutic or imaging contrast agent to diseased bone or pathological calcifications. Bisphosphonates (BPs) are the most widely utilized bone-targeting ligand due to exhibiting high binding affinity to hydroxyapatite mineral. BPs can be conjugated to an agent that would otherwise have little or no affinity for the sites of interest. This article summarizes the current state of knowledge and practice for the use of BPs as ligands for targeted delivery to bone and mineral deposits. The clinical history of BPs is briefly summarized to emphasize the success of these molecules as therapeutics for metabolic bone diseases. Mechanisms of binding and the relative binding affinity of various BPs to bone mineral are introduced, including common methods for measuring binding affinity in vitro and in vivo. Current research is highlighted for the use of BP ligands for targeted delivery of BP conjugates in various applications, including (1) therapeutic drug delivery for metabolic bone diseases, bone cancer, other bone diseases, and engineered drug delivery platforms; (2) imaging probes for scintigraphy, fluorescence, positron emission tomography, magnetic resonance imaging, and computed tomography; and (3) radiotherapy. Last, and perhaps most importantly, key structure-function relationships are considered for the design of drugs with BP ligands, including the tether length between the BP and drug, the size of the drug, the number of BP ligands per drug, cleavable tethers between the BP and drug, and conjugation schemes.
Collapse
Affiliation(s)
- Lisa E Cole
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Tracy Vargo-Gogola
- Department of Biochemistry and Molecular Biology, Indiana University Simon Cancer Center, Indiana University School of Medicine-South Bend, South Bend, IN 46617, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Ryan K Roeder
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
22
|
Tewes F, Gobbo OL, Ehrhardt C, Healy AM. Amorphous Calcium Carbonate Based-Microparticles for Peptide Pulmonary Delivery. ACS APPLIED MATERIALS & INTERFACES 2016; 8:1164-1175. [PMID: 26692360 DOI: 10.1021/acsami.5b09023] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Amorphous calcium carbonate (ACC) is known to interact with proteins, for example, in biogenic ACC, to form stable amorphous phases. The control of amorphous/crystalline and inorganic/organic ratios in inhalable calcium carbonate microparticles may enable particle properties to be adapted to suit the requirements of dry powders for pulmonary delivery by oral inhalation. For example, an amorphous phase can immobilize and stabilize polypeptides in their native structure and amorphous and crystalline phases have different mechanical properties. Therefore, inhalable composite microparticles made of inorganic (i.e., calcium carbonate and calcium formate) and organic (i.e., hyaluronan (HA)) amorphous and crystalline phases were investigated for peptide and protein pulmonary aerosol delivery. The crystalline/amorphous ratio and polymorphic form of the inorganic component was altered by changing the microparticle drying rate and by changing the ammonium carbonate and HA initial concentration. The bioactivity of the model peptide, salmon calcitonin (sCT), coprocessed with alpha-1-antitrypsin (AAT), a model protein with peptidase inhibitor activity, was maintained during processing and the microparticles had excellent aerodynamic properties, making them suitable for pulmonary aerosol delivery. The bioavailability of sCT after aerosol delivery as sCT and AAT-loaded composite microparticles to rats was 4-times higher than that of sCT solution.
Collapse
Affiliation(s)
- Frederic Tewes
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin , Dublin 2, Ireland
- INSERM U 1070, Pôle Biologie-Santé, Université de Poitiers , Faculté de Médecine & Pharmacie, 1 Rue Georges Bonnet, 86022 Poitiers Cedex, France
| | - Oliviero L Gobbo
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin , Dublin 2, Ireland
| | - Carsten Ehrhardt
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin , Dublin 2, Ireland
| | - Anne Marie Healy
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin , Dublin 2, Ireland
| |
Collapse
|
23
|
Lopez-Medina E, Fan D, Coughlin LA, Ho EX, Lamont IL, Reimmann C, Hooper LV, Koh AY. Candida albicans Inhibits Pseudomonas aeruginosa Virulence through Suppression of Pyochelin and Pyoverdine Biosynthesis. PLoS Pathog 2015; 11:e1005129. [PMID: 26313907 PMCID: PMC4552174 DOI: 10.1371/journal.ppat.1005129] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 08/04/2015] [Indexed: 01/09/2023] Open
Abstract
Bacterial-fungal interactions have important physiologic and medical ramifications, but the mechanisms of these interactions are poorly understood. The gut is host to trillions of microorganisms, and bacterial-fungal interactions are likely to be important. Using a neutropenic mouse model of microbial gastrointestinal colonization and dissemination, we show that the fungus Candida albicans inhibits the virulence of the bacterium Pseudomonas aeruginosa by inhibiting P. aeruginosa pyochelin and pyoverdine gene expression, which plays a critical role in iron acquisition and virulence. Accordingly, deletion of both P. aeruginosa pyochelin and pyoverdine genes attenuates P. aeruginosa virulence. Heat-killed C. albicans has no effect on P. aeruginosa, whereas C. albicans secreted proteins directly suppress P. aeruginosa pyoverdine and pyochelin expression and inhibit P. aeruginosa virulence in mice. Interestingly, suppression or deletion of pyochelin and pyoverdine genes has no effect on P. aeruginosa’s ability to colonize the GI tract but does decrease P. aeruginosa’s cytotoxic effect on cultured colonocytes. Finally, oral iron supplementation restores P. aeruginosa virulence in P. aeruginosa and C. albicans colonized mice. Together, our findings provide insight into how a bacterial-fungal interaction can modulate bacterial virulence in the intestine. Previously described bacterial-fungal antagonistic interactions have focused on growth inhibition or colonization inhibition/modulation, yet here we describe a novel observation of fungal-inhibition of bacterial effectors critical for virulence but not important for colonization. These findings validate the use of a mammalian model system to explore the complexities of polymicrobial, polykingdom infections in order to identify new therapeutic targets for preventing microbial disease. Pseudomonas aeruginosa and Candida albicans are two medically important human pathogens that often co-infect or co-colonize the same human niches, such as the gut. In a normal healthy host, P. aeruginosa and C. albicans can colonize the gut without any significant pathologic sequelae. But in immunocompromised hosts, both pathogens can escape the gut and cause life-threatening disseminated infections. Yet the mechanisms and pathogenic consequences of interactions between these two pathogens within a living mammalian host are not well understood. Here, we use a mouse model of P. aeruginosa and C. albicans gut co-infection to better understand the mechanisms by which C. albicans inhibits P. aeruginosa infection. C. albicans inhibits the expression of P. aeruginosa genes that are vital for iron acquisition. Accordingly, deleting these iron acquisition genes in P. aeruginosa prevents infection. Understanding how microbes interact and antagonize each other may help us identify new potential therapeutic targets for preventing or treating infections.
Collapse
Affiliation(s)
- Eduardo Lopez-Medina
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Di Fan
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Laura A. Coughlin
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Evi X. Ho
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Iain L. Lamont
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Cornelia Reimmann
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Lora V. Hooper
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- The Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Center for Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Andrew Y. Koh
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
24
|
He H, Wang P, Cai C, Yang R, Tang X. VB12-coated Gel-Core-SLN containing insulin: Another way to improve oral absorption. Int J Pharm 2015; 493:451-9. [PMID: 26253378 DOI: 10.1016/j.ijpharm.2015.08.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/08/2015] [Accepted: 08/02/2015] [Indexed: 12/21/2022]
Abstract
To improve the oral absorption of insulin, a novel carrier of Vitamin B12 (VB12) gel core solid lipid nanopaticles (Gel-Core-SLN, GCSLN) was designed with a gel core, lipid matrix and VB12-coated surface. VB12-stearate was synthesized and characterized by infrared spectroscopy (IR), nuclear magnetic resonance spectroscopy (NMR) and mass spectrometry (MS). Sol-gel conversion following ultrasonic heating and double emulsion technology were combined to implant the insulin-containing gel into solid lipid nanoparticles (SLN). The influence of the mode of administration, food, the amount of VB12-stearate and the particle size on the oral absorption of insulin incorporated in the VB12-GCSLN was investigated. The determined partition coefficient (LogP) of VB12-stearate in a dichloromethane (DCM)-water system was 3.4. This new structure of VB12-GCSLN had higher insulin encapsulation efficiency (EE) of 55.9%, a lower burst release of less than 10% in the first 2h. In vivo studies demonstrated that stronger absorption of insulin with a relative pharmacological availability (PA) of 9.31% compared with the normal insulin-loaded SLN and GCSLN and fairly stable blood glucose levels up to 12h were maintained without any sharp fluctuations. This study suggests that VB12-GCSLN containing insulin appears to be a promising nano carrier for oral delivery of biomacromolecules with relatively high pharmacological availability.
Collapse
Affiliation(s)
- Haibing He
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Puxiu Wang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Cuifang Cai
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Rui Yang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| | - Xing Tang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR China
| |
Collapse
|
25
|
Oral films as breakthrough tools for oral delivery of proteins/peptides. J Control Release 2015; 211:63-73. [DOI: 10.1016/j.jconrel.2015.05.258] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 02/07/2023]
|
26
|
Umerska A, Paluch KJ, Santos-Martinez MJ, Medina C, Corrigan OI, Tajber L. Chondroitin-based nanoplexes as peptide delivery systems – Investigations into the self-assembly process, solid-state and extended release characteristics. Eur J Pharm Biopharm 2015; 93:242-53. [DOI: 10.1016/j.ejpb.2015.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/11/2015] [Accepted: 04/13/2015] [Indexed: 10/23/2022]
|
27
|
Karsdal MA, Riis BJ, Mehta N, Stern W, Arbit E, Christiansen C, Henriksen K. Lessons learned from the clinical development of oral peptides. Br J Clin Pharmacol 2015; 79:720-32. [PMID: 25408230 PMCID: PMC4415709 DOI: 10.1111/bcp.12557] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/14/2014] [Indexed: 01/11/2023] Open
Abstract
The oral delivery of peptides and proteins has been hampered by an array of obstacles. However, several promising novel oral delivery systems have been developed. This paper reviews the most advanced oral formulation technologies, and highlights key lessons and implications from studies undertaken to date with these oral formulations. Special interest is given to oral salmon calcitonin (CT), glucagon-like peptide-1 (GLP-1), insulin, PYY-(3-36), recombinant human parathyroid hormone (rhPTH(1-31)-NH2) and PTH(1-34), by different technologies. The issues addressed include (i) interaction with water, (ii) interaction with food, (iii) diurnal variation, (iv) inter- and intra-subject variability, (v) correlation between efficacy and exposure and (vi) key deliverables of different technologies. These key lessons may aid research in the development of other oral formulations.
Collapse
|
28
|
Investigation of coco-glucoside as a novel intestinal permeation enhancer in rat models. Eur J Pharm Biopharm 2014; 88:856-65. [DOI: 10.1016/j.ejpb.2014.10.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 01/28/2023]
|
29
|
Abstract
Delivery of peptides by the oral route greatly appeals due to commercial, patient convenience and scientific arguments. While there are over 60 injectable peptides marketed worldwide, and many more in development, most delivery strategies do not yet adequately overcome the barriers to oral delivery. Peptides are sensitive to chemical and enzymatic degradation in the intestine, and are poorly permeable across the intestinal epithelium due to sub-optimal physicochemical properties. A successful oral peptide delivery technology should protect potent peptides from presystemic degradation and improve epithelial permeation to achieve a target oral bioavailability with acceptable intra-subject variability. This review provides a comprehensive up-to-date overview of the current status of oral peptide delivery with an emphasis on patented formulations that are yielding promising clinical data.
Collapse
|
30
|
Prolonged hypocalcemic effect by pulmonary delivery of calcitonin loaded poly(methyl vinyl ether maleic acid) bioadhesive nanoparticles. BIOMED RESEARCH INTERNATIONAL 2014; 2014:932615. [PMID: 24701588 PMCID: PMC3950494 DOI: 10.1155/2014/932615] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 12/08/2013] [Accepted: 01/12/2014] [Indexed: 11/17/2022]
Abstract
The purpose of the present study was to design a pulmonary controlled release system of salmon calcitonin (sCT). Therefore, poly(methyl vinyl ether maleic acid) [P(MVEMA)] nanoparticles were prepared by ionic cross-linking method using Fe2+ and Zn2+ ions. Physicochemical properties of nanoparticles were studied in vitro. The stability of sCT in the optimized nanoparticles was studied by electrophoretic gel method. Plasma calcium levels until 48 h were determined in rats as pulmonary-free sCT solution or nanoparticles (25 μg·kg−1), iv solution of sCT (5 μg·kg−1), and pulmonary blank nanoparticles. The drug remained stable during fabrication and tests on nanoparticles. The optimized nanoparticles showed proper physicochemical properties. Normalized reduction of plasma calcium levels was at least 2.76 times higher in pulmonary sCT nanoparticles compared to free solution. The duration of hypocalcemic effect of pulmonary sCT nanoparticles was 24 h, while it was just 1 h for the iv solution. There was not any significant difference between normalized blood calcium levels reduction in pulmonary drug solution and iv injection. Pharmacological activity of nanoparticles after pulmonary delivery was 65% of the iv route. Pulmonary delivery of P(MVEMA) nanoparticles of sCT enhanced and prolonged the hypocalcemic effect of the drug significantly.
Collapse
|
31
|
Welling SH, Hubálek F, Jacobsen J, Brayden DJ, Rahbek UL, Buckley ST. The role of citric acid in oral peptide and protein formulations: relationship between calcium chelation and proteolysis inhibition. Eur J Pharm Biopharm 2013; 86:544-51. [PMID: 24384069 DOI: 10.1016/j.ejpb.2013.12.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
The excipient citric acid (CA) has been reported to improve oral absorption of peptides by different mechanisms. The balance between its related properties of calcium chelation and permeation enhancement compared to a proteolysis inhibition was examined. A predictive model of CA's calcium chelation activity was developed and verified experimentally using an ion-selective electrode. The effects of CA, its salt (citrate, Cit) and the established permeation enhancer, lauroyl carnitine chloride (LCC) were compared by measuring transepithelial electrical resistance (TEER) and permeability of insulin and FD4 across Caco-2 monolayers and rat small intestinal mucosae mounted in Ussing chambers. Proteolytic degradation of insulin was determined in rat luminal extracts across a range of pH values in the presence of CA. CA's capacity to chelate calcium decreased ~10-fold for each pH unit moving from pH 6 to pH 3. CA was an inferior weak permeation enhancer compared to LCC in both in vitro models using physiological buffers. At pH 4.5 however, degradation of insulin in rat luminal extracts was significantly inhibited in the presence of 10mM CA. The capacity of CA to chelate luminal calcium does not occur significantly at the acidic pH values where it effectively inhibits proteolysis, which is its dominant action in oral peptide formulations. On account of insulin's low basal permeability, inclusion of alternative permeation enhancers is likely to be necessary to achieve sufficient oral bioavailability since this is a weak property of CA.
Collapse
Affiliation(s)
- Søren H Welling
- Diabetes Research Unit, Novo Nordisk A/S, Måløv, Denmark; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jette Jacobsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Ulrik L Rahbek
- Diabetes Research Unit, Novo Nordisk A/S, Måløv, Denmark
| | | |
Collapse
|
32
|
PREAL: prediction of allergenic protein by maximum Relevance Minimum Redundancy (mRMR) feature selection. BMC SYSTEMS BIOLOGY 2013; 7 Suppl 5:S9. [PMID: 24565053 PMCID: PMC4029432 DOI: 10.1186/1752-0509-7-s5-s9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Assessment of potential allergenicity of protein is necessary whenever transgenic proteins are introduced into the food chain. Bioinformatics approaches in allergen prediction have evolved appreciably in recent years to increase sophistication and performance. However, what are the critical features for protein's allergenicity have been not fully investigated yet. RESULTS We presented a more comprehensive model in 128 features space for allergenic proteins prediction by integrating various properties of proteins, such as biochemical and physicochemical properties, sequential features and subcellular locations. The overall accuracy in the cross-validation reached 93.42% to 100% with our new method. Maximum Relevance Minimum Redundancy (mRMR) method and Incremental Feature Selection (IFS) procedure were applied to obtain which features are essential for allergenicity. Results of the performance comparisons showed the superior of our method to the existing methods used widely. More importantly, it was observed that the features of subcellular locations and amino acid composition played major roles in determining the allergenicity of proteins, particularly extracellular/cell surface and vacuole of the subcellular locations for wheat and soybean. To facilitate the allergen prediction, we implemented our computational method in a web application, which can be available at http://gmobl.sjtu.edu.cn/PREAL/index.php. CONCLUSIONS Our new approach could improve the accuracy of allergen prediction. And the findings may provide novel insights for the mechanism of allergies.
Collapse
|
33
|
Overcoming poor permeability: translating permeation enhancers for oral peptide delivery. DRUG DISCOVERY TODAY. TECHNOLOGIES 2013; 9:e71-e174. [PMID: 24064271 DOI: 10.1016/j.ddtec.2011.11.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
34
|
Mucoadhesive intestinal devices for oral delivery of salmon calcitonin. J Control Release 2013; 172:753-62. [PMID: 24035976 DOI: 10.1016/j.jconrel.2013.09.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/24/2013] [Accepted: 09/02/2013] [Indexed: 12/25/2022]
Abstract
One of the major challenges faced by therapeutic polypeptides remains their invasive route of delivery. Oral administration offers a potential alternative to injections; however, this route cannot be currently used for peptides due to their limited stability in the stomach and poor permeation across the intestine. Here, we report mucoadhesive devices for oral delivery that are inspired by the design of transdermal patches and demonstrate their capabilities in vivo for salmon calcitonin (sCT). The mucoadhesive devices were prepared by compressing a polymeric matrix containing carbopol, pectin and sodium carboxymethylcellulose (1:1:2), and were coated on all sides but one with an impermeable and flexible ethyl cellulose (EC) backing layer. Devices were tested for in vitro dissolution, mucoadhesion to intestinal mucosa, enhancement of drug absorption in vitro (Caco-2 monolayer transport) and in vivo in rats. Devices showed steady drug release with ≈75% cumulative drug released in 5h. Devices also demonstrated strong mucoadhesion to porcine small intestine to withstand forces up to 100 times their own weight. sCT-loaded mucoadhesive devices exhibited delivery of sCT across Caco-2 monolayers and across the intestinal epithelium in vivo in rats. A ≈52-fold (pharmacokinetic) and ≈44-fold (pharmacological) enhancement of oral bioavailability was observed with mucoadhesive devices when compared to direct intestinal injections. Oral delivery of devices in enteric coated capsules resulted in significant bioavailability enhancement.
Collapse
|
35
|
Wang J, Yu Y, Zhao Y, Zhang D, Li J. Evaluation and integration of existing methods for computational prediction of allergens. BMC Bioinformatics 2013; 14 Suppl 4:S1. [PMID: 23514097 PMCID: PMC3599076 DOI: 10.1186/1471-2105-14-s4-s1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Allergy involves a series of complex reactions and factors that contribute to the development of the disease and triggering of the symptoms, including rhinitis, asthma, atopic eczema, skin sensitivity, even acute and fatal anaphylactic shock. Prediction and evaluation of the potential allergenicity is of importance for safety evaluation of foods and other environment factors. Although several computational approaches for assessing the potential allergenicity of proteins have been developed, their performance and relative merits and shortcomings have not been compared systematically. Results To evaluate and improve the existing methods for allergen prediction, we collected an up-to-date definitive dataset consisting of 989 known allergens and massive putative non-allergens. The three most widely used allergen computational prediction approaches including sequence-, motif- and SVM-based (Support Vector Machine) methods were systematically compared using the defined parameters and we found that SVM-based method outperformed the other two methods with higher accuracy and specificity. The sequence-based method with the criteria defined by FAO/WHO (FAO: Food and Agriculture Organization of the United Nations; WHO: World Health Organization) has higher sensitivity of over 98%, but having a low specificity. The advantage of motif-based method is the ability to visualize the key motif within the allergen. Notably, the performances of the sequence-based method defined by FAO/WHO and motif eliciting strategy could be improved by the optimization of parameters. To facilitate the allergen prediction, we integrated these three methods in a web-based application proAP, which provides the global search of the known allergens and a powerful tool for allergen predication. Flexible parameter setting and batch prediction were also implemented. The proAP can be accessed at http://gmobl.sjtu.edu.cn/proAP/main.html. Conclusions This study comprehensively evaluated sequence-, motif- and SVM-based computational prediction approaches for allergens and optimized their parameters to obtain better performance. These findings may provide helpful guidance for the researchers in allergen-prediction. Furthermore, we integrated these methods into a web application proAP, greatly facilitating users to do customizable allergen search and prediction.
Collapse
Affiliation(s)
- Jing Wang
- Bor Luh Food Safety Center, National Center for Molecular Characterization of Genetically Modified Organisms, State Key Laboratory of Hybrid Rice, School of Life Science and Biotechnology, Shanghai Jiao Tong University, China
| | | | | | | | | |
Collapse
|
36
|
Karsdal MA, Byrjalsen I, Henriksen K, Riis BJ, Christiansen C. A Pharmacokinetic and Pharmacodynamic Comparison of Synthetic and Recombinant Oral Salmon Calcitonin. J Clin Pharmacol 2013; 49:229-34. [DOI: 10.1177/0091270008329552] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Karsdal MA, Henriksen K, Bay-Jensen AC, Molloy B, Arnold M, John MR, Byrjalsen I, Azria M, Riis BJ, Qvist P, Christiansen C. Lessons Learned From the Development of Oral Calcitonin: The First Tablet Formulation of a Protein in Phase III Clinical Trials. J Clin Pharmacol 2013; 51:460-71. [DOI: 10.1177/0091270010372625] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
38
|
Renukuntla J, Vadlapudi AD, Patel A, Boddu SHS, Mitra AK. Approaches for enhancing oral bioavailability of peptides and proteins. Int J Pharm 2013; 447:75-93. [PMID: 23428883 DOI: 10.1016/j.ijpharm.2013.02.030] [Citation(s) in RCA: 427] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/28/2012] [Accepted: 02/12/2013] [Indexed: 12/20/2022]
Abstract
Oral delivery of peptide and protein drugs faces immense challenge partially due to the gastrointestinal (GI) environment. In spite of considerable efforts by industrial and academic laboratories, no major breakthrough in the effective oral delivery of polypeptides and proteins has been accomplished. Upon oral administration, gastrointestinal epithelium acts as a physical and biochemical barrier for absorption of proteins resulting in low bioavailability (typically less than 1-2%). An ideal oral drug delivery system should be capable of (a) maintaining the integrity of protein molecules until it reaches the site of absorption, (b) releasing the drug at the target absorption site, where the delivery system appends to that site by virtue of specific interaction, and (c) retaining inside the gastrointestinal tract irrespective of its transitory constraints. Various technologies have been explored to overcome the problems associated with the oral delivery of macromolecules such as insulin, gonadotropin-releasing hormones, calcitonin, human growth factor, vaccines, enkephalins, and interferons, all of which met with limited success. This review article intends to summarize the physiological barriers to oral delivery of peptides and proteins and novel pharmaceutical approaches to circumvent these barriers and enhance oral bioavailability of these macromolecules.
Collapse
Affiliation(s)
- Jwala Renukuntla
- Division of Pharmaceutical Sciences, South College School of Pharmacy, 400 Goody's Lane, Knoxville, TN 37931, USA
| | | | | | | | | |
Collapse
|
39
|
Colonic absorption of salmon calcitonin using tetradecyl maltoside (TDM) as a permeation enhancer. Eur J Pharm Sci 2013; 48:726-34. [PMID: 23354154 DOI: 10.1016/j.ejps.2013.01.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/14/2012] [Accepted: 01/09/2013] [Indexed: 11/21/2022]
Abstract
Calcitonin is used as a second line treatment of postmenopausal osteoporosis, but widespread acceptance is somewhat limited by subcutaneous and intranasal routes of delivery. This study attempted to enable intestinal sCT absorption in rats using the mild surfactant, tetradecyl maltoside (TDM) as an intestinal permeation enhancer. Human Caco-2 and HT29-MTX-E12 mucus-covered intestinal epithelial monolayers were used for permeation studies. Rat in situ intestinal instillation studies were conducted to evaluate the absorption of sCT with and without 0.1 w/v% TDM in jejunum, ileum and colon. TDM significantly enhanced sCT permeation across intestinal epithelial monolayers, most likely due to combined paracellular and transcellular actions. In situ, TDM caused an increased absolute bioavailability of sCT in rat colon from 1.0% to 4.6%, whereas no enhancement increase was observed in ileal and jejunal instillations. Histological analysis suggested mild perturbation of colonic epithelia in segments instilled with sCT and TDM. These data suggest that the membrane composition of the colon is different to the small intestine and that it is more amenable to permeation enhancement. Thus, formulations designed to release payload in the colon could be advantageous for systemic delivery of poorly permeable molecules.
Collapse
|
40
|
Chen C, Fan T, Jin Y, Zhou Z, Yang Y, Zhu X, Zhang ZR, Zhang Q, Huang Y. Orally delivered salmon calcitonin-loaded solid lipid nanoparticles prepared by micelle-double emulsion method via the combined use of different solid lipids. Nanomedicine (Lond) 2012; 8:1085-100. [PMID: 23075315 DOI: 10.2217/nnm.12.141] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM The purpose of this study was to develop a new orally delivered nanoparticulate system to improve the bioavailability of salmon calcitonin (sCT). MATERIALS & METHODS Four sCT-loaded solid lipid nanoparticles (SLNs) were prepared successfully by micelle-double emulsion technique via either the sole use of stearic acid (SA) or the combined use of SA and triglycerides (including tripalmitin [TP], trimyristin or trilaurin). RESULTS Compared with other SLNs, the combination of SA and TP could not only significantly improve the colloidal stability of SLNs and enhance the drug stability in the simulated intestinal fluids, but also intensively increase the intracellular uptake of drugs compared with the other SLNs (p < 0.05). The mechanism of internalization was an active transport involved in clathrin- and caveolae-dependent endocytosis. In vivo, the sCT SLNs prepared with SA and TP exhibited the highest reduction of plasma Ca(2+) level (17.44 ± 3.68%) with a bioavailability of 13.01 ± 3.24%. CONCLUSION The SLNs formed by SA and TP as the solid lipids may be a promising carrier for oral delivery of peptide drugs.
Collapse
Affiliation(s)
- Chunhui Chen
- Key Laboratory of Drug Targeting & Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Number 17 Block 3 Southern Renmin Road, Chengdu, China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Li N, Li XR, Zhou YX, Li WJ, Zhao Y, Ma SJ, Li JW, Gao YJ, Liu Y, Wang XL, Yin DD. The use of polyion complex micelles to enhance the oral delivery of salmon calcitonin and transport mechanism across the intestinal epithelial barrier. Biomaterials 2012; 33:8881-92. [PMID: 22975427 DOI: 10.1016/j.biomaterials.2012.08.047] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 08/22/2012] [Indexed: 01/19/2023]
Abstract
The objective of the present study was to demonstrate the effect of polyanionic copolymer mPEG-grafted-alginic acid (mPEG-g-AA)-based polyion complex (PIC) micelles on enhancing the oral absorption of salmon calcitonin (sCT) in vivo and in vitro and identify the transepithelial transport mechanism of PIC micelles across the intestinal barrier. mPEG-g-AA was first successfully synthesized and characterized in cytotoxicity. The PIC micelles were approximately of 72 nm in diameter with a narrow distribution. The extremely significant enhancement of hypocalcemia efficacy of sCT-loaded PIC micelles in rats was evidenced by intraduodenal administration in comparison with sCT solution. The presence of mPEG-grafted-chitosan in PIC micelles had no favorable effect on this action in the referred content. In the Caco-2 transport studies, PIC micelles could significantly increase the permeability of sCT across Caco-2 monolayers without significantly affecting transepithelial electrical resistance values during the transport study. No evident alterations in the F-actin cytoskeleton were detected by confocal microscope observation following treatment of the cell monolayers with PIC micelles, which further certified the incapacity of PIC micelles to open the intercellular tight junctions. In addition, TEM observations showed that the intact PIC micelles were transported across the everted gut sac. These suggested that the transport of PIC micelles across Caco-2 cell monolayers involve a predominant transcytosis mechanism via endocytosis rather than paracellular pathway. Furthermore, PIC micelles were localized in both the cytoplasm and the nuclei observed by CLSM. Therefore, PIC micelles might be a potentially applicable tool for enhancing the oral absorption of cationic peptide and protein drugs.
Collapse
Affiliation(s)
- Na Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
In spite of the numerous barriers inherent in the oral delivery of therapeutically active proteins, research into the development of functional protein-delivery systems is still intense. The effectiveness of such oral protein-delivery systems depend on their ability to protect the incorporated protein from proteolytic degradation in the GI tract and enhance its intestinal absorption without significantly compromising the bioactivity of the protein. Among these delivery systems are polyelectrolyte complexes (PECs) which are composed of polyelectrolyte polymers complexed with a protein via coulombic and other interactions. This review will focus on the current status of PECs with a particular emphasis on the potential and limitations of multi- or inter-polymer PECs used to facilitate oral protein delivery.
Collapse
|
43
|
Fan Y, Li X, Zhou Y, Fan C, Wang X, Huang Y, Liu Y. Improved intestinal delivery of salmon calcitonin by water-in-oil microemulsions. Int J Pharm 2011; 416:323-30. [DOI: 10.1016/j.ijpharm.2011.06.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 06/15/2011] [Accepted: 06/17/2011] [Indexed: 10/18/2022]
|
44
|
Ma B, Zha H, Li N, Yang D, Lin G. Effect of Structural Modification of α-Aminoxy Peptides on Their Intestinal Absorption and Transport Mechanism. Mol Pharm 2011; 8:1073-82. [DOI: 10.1021/mp1003866] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Bin Ma
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Huiyan Zha
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR
| | - Na Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Dan Yang
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR
| | - Ge Lin
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| |
Collapse
|
45
|
Mitra P, Audus K, Williams G, Yazdanian M, Galinis D. A comprehensive study demonstrating that p-glycoprotein function is directly affected by changes in pH: implications for intestinal pH and effects on drug absorption. J Pharm Sci 2011; 100:4258-68. [PMID: 21538355 DOI: 10.1002/jps.22596] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 03/11/2011] [Accepted: 04/12/2011] [Indexed: 02/03/2023]
Abstract
The purpose of this study was to investigate whether changes in the pH of the gastrointestinal tract can directly affect P-glycoprotein (P-gp) function. The effect of changes in extracellular pH on P-gp functionality was examined by testing colchicine (a nonionizable P-gp substrate) in bidirectional Caco-2 and MDR1-Madine Darby canine kidney (MDCK) cell permeability assays, in which the pH of the apical and basolateral chambers was varied. Reduction of the pH from 7.4 to 5.0 and 4.5 markedly increased the apical-to-basolateral flux of colchicine and reduced the basolateral-to-apical flux. The efflux ratio for colchicine was reduced to 1.2 at pH 4.5, compared with values greater than 20 that were measured in the pH range of 5.5-7.4. A similar result was obtained when MDR1-MDCK cells were used in the bidirectional permeability studies. Other nonionizable P-gp substrates (digoxin, dexamethasone, paclitaxel, and etoposide) responded to acidic pH (4.5) in a manner similar to colchicine. Reduced P-gp ATPase activity is a reason for the diminished P-gp function observed at pH 4.5.
Collapse
Affiliation(s)
- Pallabi Mitra
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas
| | | | | | | | | |
Collapse
|
46
|
|
47
|
Andrade F, Videira M, Ferreira D, Sarmento B. Nanocarriers for pulmonary administration of peptides and therapeutic proteins. Nanomedicine (Lond) 2011; 6:123-41. [DOI: 10.2217/nnm.10.143] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Peptides and therapeutic proteins have been the target of intense research and development in recent years by the pharmaceutical and biotechnology industry. Preferably, they are administered through the parenteral route, which is associated with reduced patient compliance. Formulations for noninvasive administration of peptides and therapeutic proteins are currently being developed. Among them, inhalation appears as a promising alternative for the administration of such products. Several formulations for pulmonary delivery are in various stages of development. Despite positive results, conventional formulations have some limitations such as reduced bioavailability and side effects. Nanocarriers may be an alternative way to overcome the problems of conventional formulations. Some nanocarrier-based formulations of peptides and therapeutic proteins are currently under development. The results obtained are promising, revealing the usefulness of these systems in the delivery of such drugs.
Collapse
Affiliation(s)
- Fernanda Andrade
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua Aníbal Cunha 164 4050-047, Portugal
| | - Mafalda Videira
- iMed.UL – Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Portugal
| | - Domingos Ferreira
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua Aníbal Cunha 164 4050-047, Portugal
| | - Bruno Sarmento
- Centro de Investigação em Ciências da Saúde (CICS), Department of Pharmaceutical Sciences, Instituto Superior de Ciências da Saúde – Norte, Gandra, Portugal
| |
Collapse
|
48
|
Henriksen K, Bay-Jensen AC, Christiansen C, Karsdal MA. Oral salmon calcitonin--pharmacology in osteoporosis. Expert Opin Biol Ther 2010; 10:1617-29. [PMID: 20932224 DOI: 10.1517/14712598.2010.526104] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
IMPORTANCE OF THE FIELD Osteoporosis is a slow progressive disease with develops over decades, and where intervention is needed for an extended number of years. This highlights the need for safe intervention possibilities, which have sustained beneficial effects post-treatment. AREAS COVERED IN THIS REVIEW Articles on salmon calcitonin appearing on Pubmed from 1960 until today, with focus on a newly developed oral formulation showing increased exposure and efficacy compared with nasal formulation is reviewed. The second half focuses on long-term phenomena, such as bone quality and resolution effects. The final part discusses potential additional benefits of salmon calcitonin. WHAT THE READER WILL GAIN Insight into the clinical development of an orally formulated peptide, as well as a detailed understanding of why this approach could revive salmon calcitonin as a treatment for osteoporosis. TAKE HOME MESSAGE The oral formulation of salmon calcitonin provides additional benefits and increased efficacy on bone based on Phase I and II clinical trials data, as compared with the nasal formulation. Hence, the results on the ongoing Phase III fracture trial are awaited with great interest.
Collapse
Affiliation(s)
- Kim Henriksen
- Nordic Bioscience A/S, Herlev Hovedgade 207, DK-2730 Herlev, Denmark.
| | | | | | | |
Collapse
|
49
|
Synthesis, characterization and in vitro evaluation of a bone targeting delivery system for salmon Calcitonin. Int J Pharm 2010; 394:26-34. [DOI: 10.1016/j.ijpharm.2010.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 04/05/2010] [Accepted: 04/13/2010] [Indexed: 11/18/2022]
|
50
|
Wu L, Zhang G, Lu Q, Sun Q, Wang M, Li N, Gao Z, Sun Y, Li T, Han D, Yu X, Wang L, Sun W, Zhao D, Wu Y, Lu Y, Chen X. Evaluation of salmon calcitonin (sCT) enteric-coated capsule for enhanced absorption and GI tolerability in rats. Drug Dev Ind Pharm 2010. [DOI: 10.3109/03639040903173580] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|