1
|
Song KH. Effect of Tight Junction-Modulating FCIGRL-Modified Peptides on the Intestinal Absorption of Doxorubicin in Rats. Pharmaceutics 2024; 16:650. [PMID: 38794312 PMCID: PMC11125019 DOI: 10.3390/pharmaceutics16050650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Doxorubicin is a potent chemotherapy drug, but its oral bioavailability is limited due to its low membrane permeability. Thus, absorption enhancers such as zonula occludens toxin and its six-mer fragment, FCIGRL, have been studied to address this issue. This study aimed to evaluate the effectiveness of four peptides (Pep1, Pep2, Pep3, and Pep4) derived from FCIGRL and investigate the changes in the absorption of doxorubicin, to propose an absorption enhancer for doxorubicin. Pep1 is a modified version of FCIGRL in which the hydroxyl group at the C-terminus is replaced with an amino group. Pep2 is a modified Pep1 in which cysteine is replaced with N3-substituted dipropionic acid. Pep3 and Pep4 are Pep2-modified homodimers. Pharmacokinetic analysis was performed in rats after the intraduodenal administration of doxorubicin solutions containing each FCIGRL-modified peptide and the stabilizer levan or benzalkonium chloride (BC). The results showed that Pep3 and Pep4 administered with levan each significantly increased the intestinal absorption of doxorubicin, as did Pep2 administered with levan/BC. In particular, 10 mg·kg-1 of Pep4 with levan significantly increased the area under the curve (AUC)0-240min of doxorubicin by 2.38-fold (p < 0.01) and the peak concentration (Cmax) by 3.30-fold (p < 0.01) compared to the control solution. The study findings indicate that Pep2, Pep3, and primarily Pep4 are novel absorption enhancers that can open tight junctions for doxorubicin, and the effectiveness of the peptides was directly affected by the presence of levan or levan/BC.
Collapse
Affiliation(s)
- Keon-Hyoung Song
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soonchunhyang University, Asan 31538, Republic of Korea
| |
Collapse
|
2
|
Zorea A, Pellow D, Levin L, Pilosof S, Friedman J, Shamir R, Mizrahi I. Plasmids in the human gut reveal neutral dispersal and recombination that is overpowered by inflammatory diseases. Nat Commun 2024; 15:3147. [PMID: 38605009 PMCID: PMC11009399 DOI: 10.1038/s41467-024-47272-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Plasmids are pivotal in driving bacterial evolution through horizontal gene transfer. Here, we investigated 3467 human gut microbiome samples across continents and disease states, analyzing 11,086 plasmids. Our analyses reveal that plasmid dispersal is predominantly stochastic, indicating neutral processes as the primary driver of their wide distribution. We find that only 20-25% of plasmid DNA is being selected in various disease states, constraining its distribution across hosts. Selective pressures shape specific plasmid segments with distinct ecological functions, influenced by plasmid mobilization lifestyle, antibiotic usage, and inflammatory gut diseases. Notably, these elements are more commonly shared within groups of individuals with similar health conditions, such as Inflammatory Bowel Disease (IBD), regardless of geographic location across continents. These segments contain essential genes such as iron transport mechanisms- a distinctive gut signature of IBD that impacts the severity of inflammation. Our findings shed light on mechanisms driving plasmid dispersal and selection in the human gut, highlighting their role as carriers of vital gene pools impacting bacterial hosts and ecosystem dynamics.
Collapse
Affiliation(s)
- Alvah Zorea
- National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, 8410501, Be'er Sheva, Israel
- Department of Life Sciences, Ben-Gurion University of the Negev, 8410501, Be'er Sheva, Israel
- The Goldman Sonnenfeldt School of Sustainability and Climate Change, Ben-Gurion University of the Negev, 8410501, Be'er Sheva, Israel
| | - David Pellow
- Blavatnik School of Computer Science, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Liron Levin
- Bioinformatics Core Facility, llse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501, Be'er Sheva, Israel
| | - Shai Pilosof
- Department of Life Sciences, Ben-Gurion University of the Negev, 8410501, Be'er Sheva, Israel
- The Goldman Sonnenfeldt School of Sustainability and Climate Change, Ben-Gurion University of the Negev, 8410501, Be'er Sheva, Israel
| | - Jonathan Friedman
- Institute of Environmental Sciences, Hebrew University, Rehovot, Israel
| | - Ron Shamir
- Blavatnik School of Computer Science, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Itzhak Mizrahi
- National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, 8410501, Be'er Sheva, Israel.
- Department of Life Sciences, Ben-Gurion University of the Negev, 8410501, Be'er Sheva, Israel.
- The Goldman Sonnenfeldt School of Sustainability and Climate Change, Ben-Gurion University of the Negev, 8410501, Be'er Sheva, Israel.
| |
Collapse
|
3
|
Jha SK, Imran M, Jha LA, Hasan N, Panthi VK, Paudel KR, Almalki WH, Mohammed Y, Kesharwani P. A Comprehensive review on Pharmacokinetic Studies of Vaccines: Impact of delivery route, carrier-and its modulation on immune response. ENVIRONMENTAL RESEARCH 2023; 236:116823. [PMID: 37543130 DOI: 10.1016/j.envres.2023.116823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
The lack of knowledge about the absorption, distribution, metabolism, and excretion (ADME) of vaccines makes former biopharmaceutical optimization difficult. This was shown during the COVID-19 immunization campaign, where gradual booster doses were introduced.. Thus, understanding vaccine ADME and its effects on immunization effectiveness could result in a more logical vaccine design in terms of formulation, method of administration, and dosing regimens. Herein, we will cover the information available on vaccine pharmacokinetics, impacts of delivery routes and carriers on ADME, utilization and efficiency of nanoparticulate delivery vehicles, impact of dose level and dosing schedule on the therapeutic efficacy of vaccines, intracellular and endosomal trafficking and in vivo fate, perspective on DNA and mRNA vaccines, new generation sequencing and mathematical models to improve cancer vaccination and pharmacology, and the reported toxicological study of COVID-19 vaccines. Altogether, this review will enhance the reader's understanding of the pharmacokinetics of vaccines and methods that can be implied in delivery vehicle design to improve the absorption and distribution of immunizing agents and estimate the appropriate dose to achieve better immunogenic responses and prevent toxicities.
Collapse
Affiliation(s)
- Saurav Kumar Jha
- Department of Biomedicine, Health & Life Convergence Sciences, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea; Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur, 208016, Uttar Pradesh, India.
| | - Mohammad Imran
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Laxmi Akhileshwar Jha
- H. K. College of Pharmacy, Mumbai University, Pratiksha Nagar, Jogeshwari, West Mumbai, 400102, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Vijay Kumar Panthi
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Science, Centenary Institute and University of Technology Sydney, Sydney, 2007, Australia
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 24381, Saudi Arabia
| | - Yousuf Mohammed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
4
|
Taverner A, Almansour K, Gridley K, Marques ARL, MacKay J, Eggleston IM, Mrsny RJ. Structure-function analysis of tight junction-directed permeation enhancer PIP250. J Control Release 2023; 364:S0168-3659(23)00705-8. [PMID: 39491173 DOI: 10.1016/j.jconrel.2023.10.044] [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: 09/04/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/05/2024]
Abstract
The intestinal paracellular route of absorption is modulated via tight junction (TJ) structures located at the apical neck of polarized intestinal epithelial cells to restrict solute movement through the intercellular space between them. Tight junctions open or close in response to changes in the phosphorylation status of light chain (MLC) at position Ser-19. This phosphorylation event is primarily controlled by MLC kinase (MLCK) and MLC phosphatase (MLCP), the latter being a holoenzyme that involves interaction between protein phosphatase 1 (PP1) and myosin targeting protein 1 (MYPT1). An entirely D-amino acid Permeant Inhibitor of Phosphatase (PIP) peptide sequence designed to disrupt PP1-MYPT1 interactions at the cytoplasmic surface of TJs, PIP250 (rrfkvktkkrk) localized at intracellular TJ structures, altered expression levels of specific TJ proteins, increased cellular phosphorylated MLC (pMLC) levels, binding to PP1, decreased epithelial barrier function, and significantly increased systemic uptake of the poorly absorbed antibiotic gentamicin in vivo. A series of PIP250 peptide analogues showed that positions phe3 and val5 were critical to its functional properties, with some providing opportunities to tune the dynamic actions of its TJ modulation properties. These data confirm the activity of PIP250 as a rationally designed oral permeation enhancer and validated key amino acids involved in its interaction with PP1 that define its overall actions; the magnitude and duration of these enhancing properties were associated with the MYPT1-mimetic properties of the PIP250 peptide analogues described.
Collapse
Affiliation(s)
- Alistair Taverner
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| | - Khaled Almansour
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Ha'il 55473, Saudi Arabia
| | - Kate Gridley
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| | - Ana Rita Lima Marques
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| | - Julia MacKay
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| | - Ian M Eggleston
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| | - Randall J Mrsny
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK.
| |
Collapse
|
5
|
Ramirez-Velez I, Belardi B. Storming the gate: New approaches for targeting the dynamic tight junction for improved drug delivery. Adv Drug Deliv Rev 2023; 199:114905. [PMID: 37271282 PMCID: PMC10999255 DOI: 10.1016/j.addr.2023.114905] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/20/2023] [Accepted: 05/29/2023] [Indexed: 06/06/2023]
Abstract
As biologics used in the clinic outpace the number of new small molecule drugs, an important challenge for their efficacy and widespread use has emerged, namely tissue penetrance. Macromolecular drugs - bulky, high-molecular weight, hydrophilic agents - exhibit low permeability across biological barriers. Epithelial and endothelial layers, for example within the gastrointestinal tract or at the blood-brain barrier, present the most significant obstacle to drug transport. Within epithelium, two subcellular structures are responsible for limiting absorption: cell membranes and intercellular tight junctions. Previously considered impenetrable to macromolecular drugs, tight junctions control paracellular flux and dictate drug transport between cells. Recent work, however, has shown tight junctions to be dynamic, anisotropic structures that can be targeted for delivery. This review aims to summarize new approaches for targeting tight junctions, both directly and indirectly, and to highlight how manipulation of tight junction interactions may help usher in a new era of precision drug delivery.
Collapse
Affiliation(s)
- Isabela Ramirez-Velez
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, United States
| | - Brian Belardi
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
6
|
Spoorthi Shetty S, Halagali P, Johnson AP, Spandana KMA, Gangadharappa HV. Oral insulin delivery: Barriers, strategies, and formulation approaches: A comprehensive review. Int J Biol Macromol 2023:125114. [PMID: 37263330 DOI: 10.1016/j.ijbiomac.2023.125114] [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: 05/03/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
Diabetes Mellitus is characterized by a hyperglycemic condition which can either be caused by the destruction of the beta cells or by the resistance developed against insulin in the cells. Insulin is a peptide hormone that regulates the metabolism of carbohydrates, proteins, and fats. Type 1 Diabetes Mellitus needs the use of Insulin for efficient management. However invasive methods of administration may lead to reduced adherence by the patients. Hence there is a need for a non-invasive method of administration. Oral Insulin has several merits over the conventional method including patient compliance, and reduced cost, and it also mimics endogenous insulin and hence reaches the liver by the portal vein at a higher concentration and thereby showing improved efficiency. However oral Insulin must pass through several barriers in the gastrointestinal tract. Some strategies that could be utilized to bypass these barriers include the use of permeation enhancers, absorption enhancers, use of suitable polymers, use of suitable carriers, and other agents. Several formulation types have been explored for the oral delivery of Insulin like hydrogels, capsules, tablets, and patches which have been described briefly by the article. A lot of attempts have been made for developing oral insulin delivery however none of them have been commercialized due to numerous shortcomings. Currently, there are several formulations from the companies that are still in the clinical phase, the success or failure of some is yet to be seen in the future.
Collapse
Affiliation(s)
- S Spoorthi Shetty
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Praveen Halagali
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Asha P Johnson
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - K M Asha Spandana
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - H V Gangadharappa
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India.
| |
Collapse
|
7
|
Fein KC, Gleeson JP, Cochran K, Lamson NG, Doerfler R, Melamed JR, Whitehead KA. Long-term daily oral administration of intestinal permeation enhancers is safe and effective in mice. Bioeng Transl Med 2023; 8:e10342. [PMID: 36684095 PMCID: PMC9842030 DOI: 10.1002/btm2.10342] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 01/25/2023] Open
Abstract
Although protein drugs are powerful biologic therapeutics, they cannot be delivered orally because their large size and hydrophilicity limit their absorption across the intestinal epithelium. One potential solution is the incorporation of permeation enhancers into oral protein formulations; however, few have advanced clinically due to toxicity concerns surrounding chronic use. To better understand these concerns, we conducted a 30-day longitudinal study of daily oral permeation enhancer use in mice and resultant effects on intestinal health. Specifically, we investigated three permeation enhancers: sodium caprate (C10), an industry standard, as well as 1-phenylpiperazine (PPZ) and sodium deoxycholate (SDC). Over 30 days of treatment, all mice gained weight, and none required removal from the study due to poor health. Furthermore, intestinal permeability did not increase following chronic use. We also quantified the gene expression of four tight junction proteins (claudin 2, claudin 3, ZO-1, and JAM-A). Significant differences in gene expression between untreated and permeation enhancer-treated mice were found, but these varied between treatment groups, with most differences resolving after a 1-week washout period. Immunofluorescence microscopy revealed no observable differences in protein localization or villus architecture between treated and untreated mice. Overall, PPZ and SDC performed comparably to C10, one of the most clinically advanced enhancers, and results suggest that the chronic use of some permeation enhancers may be therapeutically viable from a safety standpoint.
Collapse
Affiliation(s)
- Katherine C. Fein
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - John P. Gleeson
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Kyle Cochran
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Nicholas G. Lamson
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Rose Doerfler
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Jilian R. Melamed
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Kathryn A. Whitehead
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| |
Collapse
|
8
|
Shi Y, Liu L, Yin M, Zhao Z, Liang Y, Sun K, Li Y. Mucus- and pH-mediated controlled release of core-shell chitosan nanoparticles in the gastrointestinal tract for diabetes treatment. J Drug Target 2023; 31:65-73. [PMID: 35861405 DOI: 10.1080/1061186x.2022.2104296] [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: 01/05/2023]
Abstract
For the successful oral delivery of peptide drugs, considerable barriers created by the harsh environment of the gastrointestinal tract, mucus, and epithelial cells must be overcome. This study was to establish a core-shell structure with chitosan (CS) nanoparticles (NP) as the core and poly-N-(2-hydroxypropyl) methacrylamide (pHPMA) as the intelligent escape shell to overcome pH and mucus barriers and improve the delivery efficiency of peptide drugs. A core-shell system (COS) composed of pHPMA-AT-1002-cys-chitosan (LRA-PA-CNPs) was prepared and used for the treatment of type 2 diabetes mellitus with the large-molecule peptide drug liraglutide (LRA). The complete COS system was observed through electron microscopy; the particle size of the LRA-PA-CNPs was approximately 160 nm; the encapsulation efficiency was approximately 69% ± 5%; the zeta potential was close to neutral; the mucus and epithelial penetration of the COS system were increased; and animal experiments showed that the COS system enhanced the oral hypoglycaemic effect of LRA.HIGHLIGHTSIntelligent escape material of poly-N-(2-hydroxypropyl) methacrylamide as the shell.Core-shell nanoparticles penetrate the mucus layer and exposing the chitosan core.Overcome pH and mucus barriers to improve the delivery efficiency of peptide drugs.
Collapse
Affiliation(s)
- Yanan Shi
- College of Life Science, Yantai University, Yantai, China
| | - Lanze Liu
- College of Life Science, Yantai University, Yantai, China
| | - Miaomiao Yin
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Zhenyu Zhao
- College of Life Science, Yantai University, Yantai, China
| | - Yanzi Liang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China.,State Key Laboratory of Long-acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai, China
| | - Youxin Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China.,State Key Laboratory of Long-acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai, China
| |
Collapse
|
9
|
Kim D, Jin L, Park EJ, Na DH. Peptide permeation enhancers for improving oral bioavailability of macromolecules. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00609-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
10
|
Islam MR, Akash S, Rahman MM, Nowrin FT, Akter T, Shohag S, Rauf A, Aljohani AS, Simal-Gandara J. Colon cancer and colorectal cancer: Prevention and treatment by potential natural products. Chem Biol Interact 2022; 368:110170. [DOI: 10.1016/j.cbi.2022.110170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/24/2022] [Accepted: 09/03/2022] [Indexed: 11/29/2022]
|
11
|
Naryzhny S, Legina O. Zonulin — regulation of tight contacts in the brain and intestine — facts and hypotheses. BIOMEDITSINSKAYA KHIMIYA 2022; 68:309-320. [DOI: 10.18097/pbmc20226805309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In recent years, the interrelationship between the brain and the gut has become an area of high scientific interest. The intestine is responsible not only for digestion, as it contains millions of neurons, its own immune system, and affects the emotional and cognitive processes. The relationship between the gut and the brain suggests that the processes carried out by the gut microbiota play a significant role in the regulation of brain function, and vice versa. A special role here is played by intercellular tight junctions (TJ), where the zonulin protein holds an important place. Zonulin, an unprocessed precursor of mature haptoglobin, is the only physiological modulator of intercellular TJ that can reversibly regulate the permeability of the intestinal (IB) and blood-brain (BBB) barriers in the human body. BBB disruption and altered microbiota composition are associated with many diseases, including neurological disorders and neuroinflammation. That is, there is a gut-brain axis (GBA) — a communication system through which the brain modulates the functions of the gastrointestinal tract (GIT) and vice versa. GBA is based on neuronal, endocrine, and immunological mechanisms that are interconnected at the organismal, organ, cellular, and molecular levels.
Collapse
Affiliation(s)
- S.N. Naryzhny
- Institute of Biomedical Chemistry, Moscow, Russia; Petersburg Institute of Nuclear Physics B.P. Konstantinova National Research Center “Kurchatov Institute”, Gatchina, Russia
| | - O.K. Legina
- Petersburg Institute of Nuclear Physics B.P. Konstantinova National Research Center “Kurchatov Institute”, Gatchina, Russia
| |
Collapse
|
12
|
Novel Drug Therapeutics in Celiac Disease: A Pipeline Review. Drugs 2022; 82:1515-1526. [PMID: 36251239 DOI: 10.1007/s40265-022-01784-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 11/03/2022]
Abstract
Celiac disease (CeD) is a chronic, autoimmune systemic disorder triggered by the ingestion of gluten, a protein found in foods such as wheat, rye, and barley. The only effective treatment for CeD is complete removal of gluten from the diet. A strict gluten-free diet (GFD) results in symptomatic, serologic, and histologic remission in most patients. However, GFD may fail to induce clinical or histologic improvement and some patients may alternatively have difficulty strictly adhering to the GFD for other reasons. Despite this, there are currently no FDA-approved drugs for the treatment of CeD. The complex pathogenic process of CeD is becoming increasingly studied and better understood, enabling the identification of various targets for future therapies. Mechanisms under evaluation include probiotics, digestion of peptides, gluten sensitization, tight junction modulation, deamidation, and immune targets. Multiple investigational drugs are in the pipeline, and several drug candidates have entered late-phase clinical trials. Indeed, current and future studies are needed to target specific etiological mechanisms and provide an alternative to GFD alone. This review provides a broad overview of the various investigative treatment approaches for CeD, summarizing the latest progress in the pipeline.
Collapse
|
13
|
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
|
14
|
Le T, Aguilar B, Mangal JL, Acharya AP. Oral drug delivery for immunoengineering. Bioeng Transl Med 2022; 7:e10243. [PMID: 35111945 PMCID: PMC8780903 DOI: 10.1002/btm2.10243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/20/2021] [Accepted: 07/25/2021] [Indexed: 11/13/2022] Open
Abstract
The systemic pharmacotherapeutic efficacy of immunomodulatory drugs is heavily influenced by its route of administration. A few common routes for the systemic delivery of immunotherapeutics are intravenous, intraperitoneal, and intramuscular injections. However, the development of novel biomaterials, in adjunct to current progress in immunoengineering, is providing an exciting area of interest for oral drug delivery for systemic targeting. Oral immunotherapeutic delivery is a highly preferred route of administration due to its ease of administration, higher patient compliance, and increased ability to generate specialized immune responses. However, the harsh environment and slow systemic absorption, due to various biological barriers, reduces the immunotherapeutic bioavailability, and in turn prevents widespread use of oral delivery. Nonetheless, cutting edge biomaterials are being synthesized to combat these biological barriers within the gastrointestinal (GI) tract for the enhancement of drug bioavailability and targeting the immune system. For example, advancements in biomaterials and synthesized drug agents have provided distinctive methods to promote localized drug absorption for the modulation of local or systemic immune responses. Additionally, novel breakthroughs in the immunoengineering field show promise in the development of vaccine delivery systems for disease prevention as well as combating autoimmune diseases, inflammatory diseases, and cancer. This review will discuss current progress made within the field of biomaterials and drug delivery systems to enhance oral immunotherapeutic availability, and how these new delivery platforms can be utilized to deliver immunotherapeutics for resolution of immune-related diseases.
Collapse
Affiliation(s)
- Tien Le
- Chemical Engineering, School for the Engineering of Matter, Transport, and EnergyArizona State UniversityTempeArizonaUSA
| | - Brian Aguilar
- Biomedical Engineering, School of Biological and Health Systems EngineeringArizona State UniversityTempeArizonaUSA
| | - Joslyn L. Mangal
- Biological Design, School for Biological and Health Systems EngineeringArizona State UniversityTempeArizonaUSA
| | - Abhinav P. Acharya
- Chemical Engineering, School for the Engineering of Matter, Transport, and EnergyArizona State UniversityTempeArizonaUSA
- Biomedical Engineering, School of Biological and Health Systems EngineeringArizona State UniversityTempeArizonaUSA
- Biological Design, School for Biological and Health Systems EngineeringArizona State UniversityTempeArizonaUSA
- Materials Science and Engineering, School for the Engineering of Matter, Transport, and energyArizona State UniversityTempeArizonaUSA
- Biodesign Center for Immunotherapy, Vaccines and VirotherapyArizona State UniversityTempeArizonaUSA
| |
Collapse
|
15
|
Sadowska-Krępa E, Rozpara M, Rzetecki A, Bańkowski S, Żebrowska A, Pilch W. Strenuous 12-h run elevates circulating biomarkers of oxidative stress, inflammation and intestinal permeability in middle-aged amateur runners: A preliminary study. PLoS One 2021; 16:e0249183. [PMID: 33793597 PMCID: PMC8016300 DOI: 10.1371/journal.pone.0249183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 03/13/2021] [Indexed: 01/08/2023] Open
Abstract
Given the solid evidence that prolonged strenuous exercise is a cause of metabolic stress, this study sought to determine whether a 12-h run would affect total oxidant status (TOS), total oxidant capacity (TOC), total antioxidant status (TAS), high-sensitivity C-reactive protein (hs-CRP) and the biomarkers of intestinal permeability (protein fatty acid-binding proteins (I-FABP) and zonulin) in middle-aged male subjects. Ten amateur long-distance runners (aged 52.0 ± 6.2 years, body height 176.9 ± 4.9 cm, body mass 73.9 ± 6.0 kg) were enrolled in the study. The venous blood samples were collected 1 hour before and right after the run and were analyzed for the levels of TAS, TOS/TOC, hs-CRP, I-FABP and zonulin. The post-run concentrations of TOS/TOC were significantly elevated (p < 0.001), but TAS changes were not significant. Pearson's correlation coefficients calculated for the post run values of TAS and TOS/TOC were statistically significant and negative (r = -0.750, p < 0.05). Significant increases in the concentrations of hs-CRP (p < 0.001), I-FABP (p < 0.05) and zonulin (p < 0.01) were noted. The results indicate that a strenuous 12-h run disturbs the prooxidant-antioxidant balance in middle-aged men, as well as promoting inflammation and impairing intestinal permeability.
Collapse
Affiliation(s)
- Ewa Sadowska-Krępa
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Michał Rozpara
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Adam Rzetecki
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Sebastian Bańkowski
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Aleksandra Żebrowska
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Wanda Pilch
- Department of Chemistry and Biochemistry, Institute for Basic Science, University of Physical Education, Krakow, Poland
| |
Collapse
|
16
|
Khan I, Daniell H. Oral delivery of therapeutic proteins bioencapsulated in plant cells: preclinical and clinical advances. Curr Opin Colloid Interface Sci 2021; 54. [PMID: 33967586 DOI: 10.1016/j.cocis.2021.101452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Oral delivery of protein drugs (PDs) made in plant cells could revolutionize current approaches of their production and delivery. Expression of PDs reduces their production cost by elimination of prohibitively expensive fermentation, purification, cold transportation/storage, and sterile injections and increases their shelf life for several years. Ability of plant cell wall to protect PDs from digestive acids/enzymes, commensal bacteria to release PDs in gut lumen after lysis of plant cell wall and role of GALT in inducing tolerance facilitate prevention or treatment allergic, autoimmune diseases or anti-drug antibody responses. Delivery of functional proteins facilitate treatment of inherited or metabolic disorders. Recent advances in making PDs free of antibiotic resistance genes in edible plant cells, long-term storage at ambient temperature maintaining their efficacy, production in cGMP facilities, IND enabling studies for clinical advancement and FDA approval of orally delivered PDs augur well for advancing this novel drug delivery platform technology.
Collapse
Affiliation(s)
- Imran Khan
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
17
|
Prediction of the enhanced insulin absorption across a triple co-cultured intestinal model using mucus penetrating PLGA nanoparticles. Int J Pharm 2020; 585:119516. [DOI: 10.1016/j.ijpharm.2020.119516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/25/2020] [Accepted: 06/05/2020] [Indexed: 01/26/2023]
|
18
|
Asfour MH. Advanced trends in protein and peptide drug delivery: a special emphasis on aquasomes and microneedles techniques. Drug Deliv Transl Res 2020; 11:1-23. [PMID: 32337668 DOI: 10.1007/s13346-020-00746-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Proteins and peptides have a great potential as therapeutic agents; they have higher efficiency and lower toxicity, compared to chemical drugs. However, their oral bioavailability is very low; also, the transdermal peptide delivery faces absorption limitations. Accordingly, most of proteins and peptides are administered by parenteral route, but there are many problems associated with this route such as patient discomfort, especially for pediatric use. Thus, it is a great challenge to develop drug delivery systems for administration of proteins and peptides by routes other than parenteral one. This review provides an overview on recent advances adopted for protein and peptide drug delivery, focusing on oral and transdermal routes. This is followed by an emphasis on two recent approaches adopted as delivery systems for protein and peptide drugs, namely aquasomes and microneedles. Aquasomes are nanoparticles fabricated from ceramics developed to enhance proteins and peptides stability, providing an adequate residence time in circulation. It consists of ceramic core coated with poly hydroxyl oligomer, on which protein and peptide drug can be adsorbed. Aquasomes preparation, characterization, and application in protein and peptide drug delivery are discussed. Microneedles are promising transdermal approach; it involves creation of micron-sized pores in the skin for enhancing the drug delivery across the skin, as their length ranged between 150 and 1500 μm. Types of microneedles with different drug delivery mechanisms, characterization, and application in protein and peptide drug delivery are discussed. Graphical abstract.
Collapse
Affiliation(s)
- Marwa Hasanein Asfour
- Pharmaceutical Technology Department, National Research Centre, El-Buhouth Street, Dokki, Cairo, 12622, Egypt.
| |
Collapse
|
19
|
Chatterjee S, Bhushan Sharma C, Lavie CJ, Adhikari A, Deedwania P, O'keefe JH. Oral insulin: an update. MINERVA ENDOCRINOL 2020; 45. [PMID: 31347343 DOI: 10.23736/s0391-1977.19.03055-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
|
20
|
Thotakura N, Kaushik L, Kumar V, Preet S, Babu PV. Advanced Approaches of Bioactive Peptide Molecules and Protein Drug Delivery Systems. Curr Pharm Des 2019; 24:5147-5163. [PMID: 30727874 DOI: 10.2174/1381612825666190206211458] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/01/2019] [Indexed: 11/22/2022]
Abstract
Despite the fact that protein and peptide therapeutics are widely employed in the treatment of various diseases, their delivery is posing an unembellished challenge to the scientists. It was discovered that delivery of these therapeutic systems through oral route is easy with high patient compliance. However, proteolytic degradation and absorption through the mucosal epithelium are the barriers in this route. These issues can be minimized by the use of enzyme inhibitors, absorption enhancers, different carrier systems or either by direct modification. In the process of investigation, it was found that transdermal route is not posing any challenges of enzymatic degradation, but, still absorption is the limitation as the outer layer of skin acts as a barrier. To suppress the effect of the barrier and increase the rate of the absorption, various advanced technologies were developed, namely, microneedle technology, iontophoresis, electroporation, sonophoresis and biochemical enhancement. Indeed, even these molecules are targeted to the cells with the use of cell-penetrating peptides. In this review, delivery of the peptide and protein therapeutics using oral, transdermal and other routes is discussed in detail.
Collapse
Affiliation(s)
- Nagarani Thotakura
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan, India
| | - Lokesh Kaushik
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan, India
| | - Vipin Kumar
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan, India
| | - Simran Preet
- Department of Biophysics, Basic Medical Sciences Block-2, Panjab University, Sector-25, Chandigarh, India
| | - Penke Vijaya Babu
- Department of chemistry, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
21
|
Singh RS, Singh A, Batra G, Kaur H, Medhi B. Novel targets for drug discovery in celiac disease. Indian J Pharmacol 2019; 51:359-365. [PMID: 31831931 PMCID: PMC6892008 DOI: 10.4103/ijp.ijp_679_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/13/2019] [Accepted: 11/16/2019] [Indexed: 11/21/2022] Open
Abstract
Celiac disease is a lifelong, immunological disorder induced by dietary protein-gluten, in a genetically susceptible populations, resulting in different clinical manifestations, the release of antibodies, and damage to the intestinal mucosa. The only recommended therapy for the disease is to strictly follow a gluten-free diet (GFD), which is difficult to comply with. A GFD is found to be ineffective in some active Celiac disease cases. Therefore, there is an unmet need for an alternative nondietary therapeutic approach. The review focuses on the novel drug targets for Celiac disease.
Collapse
Affiliation(s)
- Rahul Soloman Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashutosh Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gitika Batra
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Hardeep Kaur
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
22
|
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: 22] [Impact Index Per Article: 3.7] [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
|
23
|
Shekar A, Ramlal S, Jeyabalaji JK, Sripathy MH. Intranasal co-administration of recombinant active fragment of Zonula occludens toxin and truncated recombinant EspB triggers potent systemic, mucosal immune responses and reduces span of E. coli O157:H7 fecal shedding in BALB/c mice. Med Microbiol Immunol 2019; 208:89-100. [PMID: 30209565 DOI: 10.1007/s00430-018-0559-9] [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: 04/27/2018] [Accepted: 08/29/2018] [Indexed: 12/17/2022]
Abstract
Escherichia coli O157:H7 with its traits such as intestinal colonization and fecal-oral route of transmission demands mucosal vaccine development. E. coli secreted protein B (EspB) is one of the key type III secretory system (TTSS) targets for mucosal candidate vaccine due to its indispensable role in the pathogenesis of E. coli O157:H7. However, mucosally administered recombinant proteins have low immunogenicity which could be overcome by the use of mucosal adjuvants. The quest for safe, potent mucosal adjuvant has recognized ΔG fragment of Zonula occludens toxin of Vibrio cholerae with such properties. ΔG enhances mucosal permeability via the paracellular route by altering epithelial tight junction structure in a reversible, ephemeral and non-toxic manner. Therefore, we tested whether recombinant ΔG intranasally co-administered with truncated EspB (EspB + ΔG) could serve as an effective mucosal adjuvant. Results showed that EspB + ΔG group induced higher systemic IgG and mucosal IgA than EspB alone. Moreover, EspB alone developed Th2 type response with IgG1/IgG2a ratio (1.64) and IL-4, IL-10 cytokines whereas that of EspB + ΔG group generated mixed Th1/Th2 type immune response evident from IgG1/IgG2a ratio (1.17) as well as IL-4, IL-10 and IFN-γ cytokine levels compared to control. Sera of EspB + ΔG group inhibited TTSS mediated haemolysis of murine RBCs more effectively compared to EspB, control group and sera of both EspB + ΔG, EspB group resulted in similar levels of efficacious reduction in E. coli O157:H7 adherence to Caco-2 cells compared to control. Moreover, vaccination with EspB + ΔG resulted in significant reduction in E. coli O157:H7 fecal shedding compared to EspB and control group in experimentally challenged streptomycin-treated mice. These results demonstrate mucosal adjuvanticity of ΔG co-administered with EspB in enhancing overall immunogenicity to reduce E. coli O157:H7 shedding.
Collapse
Affiliation(s)
- Aravind Shekar
- Department of Microbiology, Defence Food Research Laboratory, Siddartha Nagar, Mysuru, Karnataka State, 570011, India
| | - Shylaja Ramlal
- Department of Microbiology, Defence Food Research Laboratory, Siddartha Nagar, Mysuru, Karnataka State, 570011, India
| | - Joseph Kingston Jeyabalaji
- Department of Microbiology, Defence Food Research Laboratory, Siddartha Nagar, Mysuru, Karnataka State, 570011, India
| | - Murali Harishchandra Sripathy
- Department of Microbiology, Defence Food Research Laboratory, Siddartha Nagar, Mysuru, Karnataka State, 570011, India.
- , RCE Layout, Bogadi 2nd stage, Mysuru, Karnataka, 570026, India.
| |
Collapse
|
24
|
Guo Y, Li Z, Su W, Wang L, Zhu Y, Qin J. A Biomimetic Human Gut-on-a-Chip for Modeling Drug Metabolism in Intestine. Artif Organs 2018; 42:1196-1205. [PMID: 30256442 DOI: 10.1111/aor.13163] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/22/2018] [Accepted: 03/27/2018] [Indexed: 12/18/2022]
Abstract
Drug metabolism in the intestine is considered to substantially contribute to the overall first-pass metabolism, which has been neglected for a long time. It is highly desirable to develop a reliable model to evaluate drug metabolism in the intestine in vitro. In this work, we made the first attempt to develop a biomimetic human gut-on-a-chip for modeling drug metabolism in intestine. In this chip, constant flow, together with porous nitrocellulose membrane and collagen I, mimics an in vivo-like intestinal microenvironment. The Caco-2 cells grown in the chip formed a compact intestinal epithelial layer with continuous expression of the tight junction protein, ZO-1. Furthermore, higher gene expression of villin, sucrase-isomaltase, and alkaline phosphatase demonstrated that cells in the biomimetic human gut-on-a-chip device were more mature with near-physiological functions compared to the control on planar substrate. In particular, cellular metabolic activity was assessed on different substrates, indicating higher metabolic efficiency of ifosfamide and verapamil in the biomimetic human gut-on-a-chip model. Taken together, our results suggested that this biomimetic human gut-on-a-chip promoted the differentiation of intestinal cells with enhanced functionality by creating a biomimetic 3D microenvironment in vitro. It might offer a bioactive, low-cost, and flexible in vitro platform for studies on intestinal metabolism as well as preclinical drug development.
Collapse
Affiliation(s)
- Yaqiong Guo
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian
- School of Future Technology, University of Chinese Academy of Sciences, Beijing
| | - Zhongyu Li
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian
| | - Wentao Su
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian
| | - Li Wang
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian
| | - Yujuan Zhu
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian
- School of Future Technology, University of Chinese Academy of Sciences, Beijing
| | - Jianhua Qin
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian
- School of Future Technology, University of Chinese Academy of Sciences, Beijing
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
25
|
Abstract
Biologics now constitute a significant element of available medical treatments. Owing to their clinical and commercial success, biologics are a rapidly growing class and have become a dominant therapeutic modality. Although most of the successful biologics to date are drugs that bear a peptidic backbone, ranging from small peptides to monoclonal antibodies (~500 residues; 150 kDa), new biologic modalities, such as nucleotide-based therapeutics and viral gene therapies, are rapidly maturing towards widespread clinical use. Given the rise of peptides and proteins in the pharmaceutical landscape, tremendous research and development interest exists in developing less-invasive or non-invasive routes for the systemic delivery of biologics, including subcutaneous, transdermal, oral, inhalation, nasal and buccal routes. This Review summarizes the current status, latest updates and future prospects for such delivery of peptides, proteins and other biologics.
Collapse
|
26
|
Ibie C, Knott R, Thompson C. Complexation of novel thiomers and insulin to protect against in vitro enzymatic degradation – towards oral insulin delivery. Drug Dev Ind Pharm 2018; 45:67-75. [DOI: 10.1080/03639045.2018.1517776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- C.O. Ibie
- School of Pharmacy and Life Sciences, Robert Gordon University, Garthdee Road, Aberdeen, UK
| | - R.M. Knott
- School of Pharmacy and Life Sciences, Robert Gordon University, Garthdee Road, Aberdeen, UK
| | - C.J. Thompson
- School of Pharmacy and Life Sciences, Robert Gordon University, Garthdee Road, Aberdeen, UK
| |
Collapse
|
27
|
Hu Q, Luo Y. Recent advances of polysaccharide-based nanoparticles for oral insulin delivery. Int J Biol Macromol 2018; 120:775-782. [PMID: 30170057 DOI: 10.1016/j.ijbiomac.2018.08.152] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/25/2018] [Accepted: 08/26/2018] [Indexed: 12/15/2022]
Abstract
Diabetes mellitus is a highly prevalent metabolic and chronic disease affecting millions of people in the world. The most common route of insulin therapy is the subcutaneous injection due to its low bioavailability and enzymatic degradation. The search for effective and high patient compliance insulin delivery systems has been a major challenge over many decades. The polysaccharide-based nanoparticles as delivery vehicles for insulin oral administration have recently attracted substantial interests. The present review highlights the recent advances on the development of nanoparticles prepared from polysaccharides, including chitosan, alginate, dextran and glucan, for oral delivery of insulin, overcoming multiple barriers in gastrointestinal tract. The aims of this review are first to summarize the strategies that have been applied in the past 5 years to fabricate polysaccharide-based nanoparticles for insulin oral delivery, and then to provide in-depth understanding on the mechanisms by which such nanoparticles protect insulin against degradation in the digestive tract and provide sustained release to enhance mucus permeation and transepithelial transport of insulin administered via oral route.
Collapse
Affiliation(s)
- Qiaobin Hu
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Yangchao Luo
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
28
|
Vaquero L, Rodríguez-Martín L, León F, Jorquera F, Vivas S. New coeliac disease treatments and their complications. GASTROENTEROLOGIA Y HEPATOLOGIA 2018; 41:191-204. [PMID: 29422237 DOI: 10.1016/j.gastrohep.2017.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/28/2017] [Accepted: 12/14/2017] [Indexed: 02/06/2023]
Abstract
The only accepted treatment for coeliac disease is strict adherence to a gluten-free diet. This type of diet may give rise to reduced patient quality of life with economic and social repercussions. For this reason, dietary transgressions are common and may elicit intestinal damage. Several treatments aimed at different pathogenic targets of coeliac disease have been developed in recent years: modification of gluten to produce non-immunogenic gluten, endoluminal therapies to degrade gluten in the intestinal lumen, increased gluten tolerance, modulation of intestinal permeability and regulation of the adaptive immune response. This review evaluates these coeliac disease treatment lines that are being researched and the treatments that aim to control disease complications like refractory coeliac disease.
Collapse
Affiliation(s)
- Luis Vaquero
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, León, España
| | | | | | - Francisco Jorquera
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, León, España; Instituto de Biomedicina (IBIOMED), Universidad de León, León, España
| | - Santiago Vivas
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, León, España; Instituto de Biomedicina (IBIOMED), Universidad de León, León, España.
| |
Collapse
|
29
|
Harloff-Helleberg S, Nielsen LH, Nielsen HM. Animal models for evaluation of oral delivery of biopharmaceuticals. J Control Release 2017; 268:57-71. [DOI: 10.1016/j.jconrel.2017.09.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/06/2017] [Accepted: 09/15/2017] [Indexed: 12/20/2022]
|
30
|
Management of eosinophilic esophagitis and celiac disease. Curr Opin Pharmacol 2017; 37:118-125. [DOI: 10.1016/j.coph.2017.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 12/23/2022]
|
31
|
Gedawy A, Martinez J, Al-Salami H, Dass CR. Oral insulin delivery: existing barriers and current counter-strategies. J Pharm Pharmacol 2017; 70:197-213. [DOI: 10.1111/jphp.12852] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 10/24/2017] [Indexed: 12/21/2022]
Abstract
Abstract
Objectives
The chronic and progressive nature of diabetes is usually associated with micro- and macrovascular complications where failure of pancreatic β-cell function and a general condition of hyperglycaemia is created. One possible factor is failure of the patient to comply with and adhere to the prescribed insulin due to the inconvenient administration route. This review summarizes the rationale for oral insulin administration, existing barriers and some counter-strategies trialled.
Key findings
Oral insulin mimics the physiology of endogenous insulin secreted by pancreas. Following the intestinal absorption of oral insulin, it reaches the liver at high concentration via the portal vein. Oral insulin on the other hand has the potential to protect pancreatic β-cells from autoimmune destruction. Structural modification, targeting a particular tissue/receptor, and the use of innovative pharmaceutical formulations such as nanoparticles represent strategies introduced to improve oral insulin bioavailability. They showed promising results in overcoming the hurdles facing oral insulin delivery, although delivery is far from ideal.
Summary
The use of advanced pharmaceutical technologies and further research in particulate carrier system delivery predominantly nanoparticle utilization would offer useful tools in delivering insulin via the oral route which in turn would potentially improve diabetic patient compliance to insulin and the overall management of diabetes.
Collapse
Affiliation(s)
- Ahmed Gedawy
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA, Australia
| | - Jorge Martinez
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA, Australia
| | - Hani Al-Salami
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA, Australia
- Curtin Health Innovation Research Institute, Bentley, WA, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA, Australia
- Curtin Health Innovation Research Institute, Bentley, WA, Australia
| |
Collapse
|
32
|
Watari A, Kodaka M, Matsuhisa K, Sakamoto Y, Hisaie K, Kawashita N, Takagi T, Yamagishi Y, Suzuki H, Tsujino H, Yagi K, Kondoh M. Identification of claudin-4 binder that attenuates tight junction barrier function by TR-FRET-based screening assay. Sci Rep 2017; 7:14514. [PMID: 29109448 PMCID: PMC5674027 DOI: 10.1038/s41598-017-15108-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/20/2017] [Indexed: 12/14/2022] Open
Abstract
Claudins are key functional and structural components of tight junctions (TJs) in epithelial cell sheets. The C-terminal fragment of Clostridium perfringens enterotoxin (C-CPE) binds to claudin-4 and reversibly modulates intestinal TJ seals, thereby enhancing paracellular transport of solutes. However, the use of C-CPE as an absorption enhancer is limited by the molecule’s immunogenicity and manufacturing cost. Here, we developed a high-throughput screening system based on the Time-Resolved Fluorescence Resonance Energy Transfer (TR-FRET) method to identify claudin-4 binders in a library collection of 32,560 compounds. Thiostrepton, identified from the screen, decreased transepithelial electrical resistance and increased flux of 4-kDa fluorescein isothiocyanate–labelled dextran (FD-4) in Caco-2 cell monolayers, a model of intestinal epithelium. Thiostrepton changed the expression, but not the localisation, of TJ components. Treatment of rat jejunum with thiostrepton increased the absorption of FD-4 without tissue toxicity, indicating that thiostrepton is a novel claudin-4 binder that enhances intestinal permeability. The screening system may therefore be a useful tool for identifying claudin-4 binders to enhance drug absorption in mucosa.
Collapse
Affiliation(s)
- Akihiro Watari
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Miki Kodaka
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Koji Matsuhisa
- Department of Stress Protein Processing, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuta Sakamoto
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kota Hisaie
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Norihito Kawashita
- Faculty of Science and Engineering, Kindai University 3-4-1 Kowakae, Higashiosaka City, Osaka, 577-8502, Japan
| | - Tatsuya Takagi
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshiaki Yamagishi
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishi-Tokyo, 202-8585, Japan
| | - Hidehiko Suzuki
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, 567-0085, Japan
| | - Hirofumi Tsujino
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kiyohito Yagi
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
33
|
Mousavi SM, Zeinoddini M, Azizi A, Saeedinia A, Monazah A. Molecular Detection of Zonula Occludens Toxin (zot) Genes in Vibrio Cholerae O1 using PCR. RESEARCH IN MOLECULAR MEDICINE 2017. [DOI: 10.29252/rmm.5.3.37] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
34
|
Wong CY, Martinez J, Carnagarin R, Dass CR. In-vitro evaluation of enteric coated insulin tablets containing absorption enhancer and enzyme inhibitor. J Pharm Pharmacol 2017; 69:285-294. [DOI: 10.1111/jphp.12694] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/18/2016] [Indexed: 01/22/2023]
Abstract
Abstract
Objectives
The aim of this study was to develop an enteric coated insulin tablet formulation using polymers, absorption enhancer and enzyme inhibitor, which protect the tablets in acidic pH and enhance systemic bioavailability.
Methods
In this study, the influence of coating by cellulose acetate hydrogen phthalate solution and chosen excipients on Glut-4 transporter translocation in C2C12 skeletal muscle cells was examined. Following the determination of optimum number of coating layers, two dissolution buffers such as 0.01 m hydrochloric acid, pH 2, and 50 mm phosphate, pH 7.4, were employed to determine the in-vitro release of insulin.
Key findings
Insulin was protected by the coating during the dissolution process. Five (5-CL) coating layers and eight (8-CL) coating layers had minimal insulin release in hydrochloric acid, but not three (3-CL) coating layers. Glut-4 translocation in C2C12 cells was promoted by the chosen excipients. No detrimental metabolic effects were observed in these cells.
Conclusion
To date, limited studies combine the overall effectiveness of multiple excipients. Our study showed that the coated tablets have an immediate release effect in phosphate buffer. In Glut-4 translocation assay, insulin was still functional after releasing from the tablet. Such tablet formulation can be potentially beneficial to type 1 diabetes patients.
Collapse
Affiliation(s)
- Chun Y Wong
- School of Pharmacy, Curtin University, Bentley, WA, Australia
| | - Jorge Martinez
- School of Pharmacy, Curtin University, Bentley, WA, Australia
- Pharmaceutical Technology Facility, School of Pharmacy, Curtin University, Bentley, WA, Australia
| | - Revathy Carnagarin
- School of Pharmacy, Curtin University, Bentley, WA, Australia
- Curtin Biosciences Research Precinct, Bentley, WA, Australia
| | - Crispin R Dass
- School of Pharmacy, Curtin University, Bentley, WA, Australia
- Curtin Biosciences Research Precinct, Bentley, WA, Australia
| |
Collapse
|
35
|
Aoun J, Hayashi M, Sheikh IA, Sarkar P, Saha T, Ghosh P, Bhowmick R, Ghosh D, Chatterjee T, Chakrabarti P, Chakrabarti MK, Hoque KM. Anoctamin 6 Contributes to Cl- Secretion in Accessory Cholera Enterotoxin (Ace)-stimulated Diarrhea: AN ESSENTIAL ROLE FOR PHOSPHATIDYLINOSITOL 4,5-BISPHOSPHATE (PIP2) SIGNALING IN CHOLERA. J Biol Chem 2016; 291:26816-26836. [PMID: 27799301 PMCID: PMC5207189 DOI: 10.1074/jbc.m116.719823] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 10/26/2016] [Indexed: 11/06/2022] Open
Abstract
Accessory cholera enterotoxin (Ace) of Vibrio cholerae has been shown to contribute to diarrhea. However, the signaling mechanism and specific type of Cl- channel activated by Ace are still unknown. We have shown here that the recombinant Ace protein induced ICl of apical plasma membrane, which was inhibited by classical CaCC blockers. Surprisingly, an Ace-elicited rise of current was neither affected by ANO1 (TMEM16A)-specific inhibitor T16A(inh)-AO1(TAO1) nor by the cystic fibrosis transmembrane conductance regulator (CFTR) blocker, CFTR inh-172. Ace stimulated whole-cell current in Caco-2 cells. However, the apical ICl was attenuated by knockdown of ANO6 (TMEM16F). This impaired phenotype was restored by re-expression of ANO6 in Caco-2 cells. Whole-cell patch clamp recordings of ANO currents in HEK293 cells transiently expressing mouse ANO1-mCherry or ANO6-GFP confirmed that Ace induced Cl- secretion. Application of Ace produced ANO6 but not the ANO1 currents. Ace was not able to induce a [Ca2+]i rise in Caco-2 cells, but cellular abundance of phosphatidylinositol 4,5-bisphosphate (PIP2) increased. Identification of the PIP2-binding motif at the N-terminal sequence among human and mouse ANO6 variants along with binding of PIP2 directly to ANO6 in HEK293 cells indicate likely PIP2 regulation of ANO6. The biophysical and pharmacological properties of Ace stimulated Cl- current along with intestinal fluid accumulation, and binding of PIP2 to the proximal KR motif of channel proteins, whose mutagenesis correlates with altered binding of PIP2, is comparable with ANO6 stimulation. We conclude that ANO6 is predominantly expressed in intestinal epithelia, where it contributes secretory diarrhea by Ace stimulation in a calcium-independent mechanism of RhoA-ROCK-PIP2 signaling.
Collapse
Affiliation(s)
- Joydeep Aoun
- From the Molecular Pathophysiology Division, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Mikio Hayashi
- the Department of Physiology, Kansai Medical University, 5-1, Shimmachi 2, Hirakata, 573 1010 Osaka, Japan
| | - Irshad Ali Sheikh
- From the Molecular Pathophysiology Division, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Paramita Sarkar
- From the Molecular Pathophysiology Division, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Tultul Saha
- From the Molecular Pathophysiology Division, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Priyanka Ghosh
- From the Molecular Pathophysiology Division, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Rajsekhar Bhowmick
- From the Molecular Pathophysiology Division, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Dipanjan Ghosh
- the Department of Biotechnology, College of Science and Technology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700019, India, and
| | - Tanaya Chatterjee
- the Department of Biochemistry, Bose Institute, P-1/12 CIT Road, Scheme-VIIM, Kolkata 700054, India
| | - Pinak Chakrabarti
- the Department of Biochemistry, Bose Institute, P-1/12 CIT Road, Scheme-VIIM, Kolkata 700054, India
| | - Manoj K Chakrabarti
- From the Molecular Pathophysiology Division, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Kazi Mirajul Hoque
- From the Molecular Pathophysiology Division, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India,
| |
Collapse
|
36
|
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
|
37
|
Sánchez-Navarro M, Garcia J, Giralt E, Teixidó M. Using peptides to increase transport across the intestinal barrier. Adv Drug Deliv Rev 2016; 106:355-366. [PMID: 27155131 DOI: 10.1016/j.addr.2016.04.031] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/24/2016] [Accepted: 04/29/2016] [Indexed: 02/05/2023]
Abstract
The oral route is the preferred for the administration of drugs; however, it has some serious limitations. One of the main disadvantages is poor permeability across the intestinal barrier. Various approaches are currently being adopted to overcome this issue. In this review, we describe the alternatives that use peptides to enhance intestinal absorption. First, we define the various sources of peptide enhancers followed by the analysis of the absorption mechanism used. We then comment on the possible toxic effects derived from their use as permeation enhancers, as well as potential formulation strategies. Finally, the advantages and drawbacks of peptides as intestinal enhancers are examined.
Collapse
|
38
|
Tao SL, Desai TA. Micromachined Polymeric Devices for Applications in Targeted Drug Delivery. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jala.2004.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
| | - Tejal A. Desai
- Therapeutic Microtechnology Laboratory, Department of Biomedical Engineering, Boston University, Boston, MA
| |
Collapse
|
39
|
Ling X, Linglong P, Weixia D, Hong W. Protective Effects of Bifidobacterium on Intestinal Barrier Function in LPS-Induced Enterocyte Barrier Injury of Caco-2 Monolayers and in a Rat NEC Model. PLoS One 2016; 11:e0161635. [PMID: 27551722 PMCID: PMC4995054 DOI: 10.1371/journal.pone.0161635] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/09/2016] [Indexed: 12/17/2022] Open
Abstract
Zonulin protein is a newly discovered modulator which modulates the permeability of the intestinal epithelial barrier by disassembling intercellular tight junctions (TJ). Disruption of TJ is associated with neonatal necrotizing enterocolitis (NEC). It has been shown bifidobacterium could protect the intestinal barrier function and prophylactical administration of bifidobacterium has beneficial effects in NEC patients and animals. However, it is still unknown whether the zonulin is involved in the gut barrier dysfunction of NEC, and the protective mechanisms of bifidobacterium on intestinal barrier function are also not well understood. The present study aims to investigate the effects of bifidobacterium on intestinal barrier function, zonulin regulation, and TJ integrity both in LPS-induced enterocyte barrier injury of Caco-2 monolayers and in a rat NEC model. Our results showed bifidobacterium markedly attenuated the decrease in transepithelial electrical resistance and the increase in paracellular permeability in the Caco-2 monolayers treated with LPS (P < 0.01). Compared with the LPS group, bifidobacterium significantly decreased the production of IL-6 and TNF-α (P < 0.01) and suppressed zonulin release (P < 0.05). In addition, bifidobacterium pretreatment up-regulated occludin, claudin-3 and ZO-1 expression (P < 0.01) and also preserved these proteins localization at TJ compared with the LPS group. In the in vivo study, bifidobacterium decreased the incidence of NEC from 88 to 47% (P < 0.05) and reduced the severity in the NEC model. Increased levels of IL-6 and TNF-α in the ileum of NEC rats were normalized in bifidobacterium treated rats (P < 0.05). Moreover, administration of bifidobacterium attenuated the increase in intestinal permeability (P < 0.01), decreased the levels of serum zonulin (P < 0.05), normalized the expression and localization of TJ proteins in the ileum compared with animals with NEC. We concluded that bifidobacterium may protect against intestinal barrier dysfunction both in vitro and in NEC. This protective effect is associated with inhibition of proinflammatory cytokine secretion, suppression of zonulin protein release and improvement of intestinal TJ integrity.
Collapse
Affiliation(s)
- Xiang Ling
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Department of Pediatrics, Second People’s Hospital of Chengdu, Chengdu, China
| | - Peng Linglong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Du Weixia
- Department of Kidney Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Hong
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
- * E-mail:
| |
Collapse
|
40
|
Muheem A, Shakeel F, Jahangir MA, Anwar M, Mallick N, Jain GK, Warsi MH, Ahmad FJ. A review on the strategies for oral delivery of proteins and peptides and their clinical perspectives. Saudi Pharm J 2016; 24:413-428. [PMID: 27330372 PMCID: PMC4908063 DOI: 10.1016/j.jsps.2014.06.004] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 06/06/2014] [Indexed: 01/10/2023] Open
Abstract
In the modern world, a number of therapeutic proteins such as vaccines, antigens, and hormones are being developed utilizing different sophisticated biotechnological techniques like recombinant DNA technology and protein purification. However, the major glitches in the optimal utilization of therapeutic proteins and peptides by the oral route are their extensive hepatic first-pass metabolism, degradation in the gastrointestinal tract (presence of enzymes and pH-dependent factors), large molecular size and poor permeation. These problems can be overcome by adopting techniques such as chemical transformation of protein structures, enzyme inhibitors, mucoadhesive polymers and permeation enhancers. Being invasive, parenteral route is inconvenient for the administration of protein and peptides, several research endeavors have been undertaken to formulate a better delivery system for proteins and peptides with major emphasis on non-invasive routes such as oral, transdermal, vaginal, rectal, pulmonary and intrauterine. This review article emphasizes on the recent advancements made in the delivery of protein and peptides by a non-invasive (peroral) route into the body.
Collapse
Affiliation(s)
- Abdul Muheem
- Department of Pharmaceutics, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi 110062, India
| | - Faiyaz Shakeel
- Center of Excellence in Biotechnology Research (CEBR), King Saud University, Riyadh, Saudi Arab
| | | | - Mohammed Anwar
- Department of Pharmaceutics, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi 110062, India
| | - Neha Mallick
- Department of Pharmaceutics, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi 110062, India
| | - Gaurav Kumar Jain
- Department of Pharmaceutics, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi 110062, India
| | - Musarrat Husain Warsi
- Department of Pharmaceutics, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi 110062, India
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi 110062, India
| |
Collapse
|
41
|
Wong CY, Martinez J, Dass CR. Oral delivery of insulin for treatment of diabetes: status quo, challenges and opportunities. J Pharm Pharmacol 2016; 68:1093-108. [DOI: 10.1111/jphp.12607] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 06/10/2016] [Indexed: 12/12/2022]
Abstract
Abstract
Objectives
Diabetes mellitus is characterised by progressive β-cell destruction and loss of function, or loss of ability of tissues to respond to insulin. Daily subcutaneous insulin injection is standard management for people with diabetes, although patient compliance is hard to achieve due to the inconvenience of injections, so other forms of delivery are being tested, including oral administration. This review summarises the developments in oral insulin administration.
Methods
The PubMed database was consulted to compile this review comparing conventional subcutaneous injection of insulin to the desired oral delivery.
Key findings
Oral administration of insulin has potential benefits in reducing pain and chances of skin infection, improving the portal levels of insulin and avoiding side effects such as hyperinsulinemia, weight gain and hypoglycaemia. Although oral delivery of insulin is an ideal administration route for patients with diabetes, several physiological barriers have to be overcome. An expected low oral bioavailability can be attributed to its high molecular weight, susceptibility to enzymatic proteolysis and low diffusion rate across the mucin barrier.
Conclusions
Strategies for increasing the bioavailability of oral insulin include the use of enzyme inhibitors, absorption enhancers, mucoadhesive polymers and chemical modification for endogenous receptor-mediated absorption. These may help significantly increase patient compliance and disease management.
Collapse
Affiliation(s)
- Chun Y Wong
- School of Pharmacy, Curtin University, Bentley, Australia
| | - Jorge Martinez
- School of Pharmacy, Curtin University, Bentley, Australia
| | - Crispin R Dass
- School of Pharmacy, Curtin University, Bentley, Australia
- Curtin Biosciences Research Precinct, Bentley, Australia
| |
Collapse
|
42
|
McGinn B, Morrison J. Investigations into the absorption of insulin and insulin derivatives from the small intestine of the anaesthetised rat. J Control Release 2016; 232:120-30. [PMID: 27084488 DOI: 10.1016/j.jconrel.2016.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/29/2016] [Accepted: 04/01/2016] [Indexed: 02/09/2023]
|
43
|
Reversible Opening of Intercellular Junctions of Intestinal Epithelial and Brain Endothelial Cells With Tight Junction Modulator Peptides. J Pharm Sci 2016; 105:754-765. [DOI: 10.1016/j.xphs.2015.11.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 10/30/2015] [Accepted: 11/05/2015] [Indexed: 01/08/2023]
|
44
|
Müller G. Personalized Diagnosis and Therapy. DRUG DISCOVERY AND EVALUATION: PHARMACOLOGICAL ASSAYS 2016:3167-3284. [DOI: 10.1007/978-3-319-05392-9_152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
45
|
Sharma G, Sharma AR, Nam JS, Doss GPC, Lee SS, Chakraborty C. Nanoparticle based insulin delivery system: the next generation efficient therapy for Type 1 diabetes. J Nanobiotechnology 2015; 13:74. [PMID: 26498972 PMCID: PMC4619439 DOI: 10.1186/s12951-015-0136-y] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/15/2015] [Indexed: 12/31/2022] Open
Abstract
Diabetic cases have increased rapidly in recent years throughout the world. Currently, for type-1 diabetes mellitus (T1DM), multiple daily insulin (MDI) injections is the most popular treatment throughout the world. At this juncture, researchers are trying to develop different insulin delivery systems, especially through oral and pulmonary route using nanocarrier based delivery system. This next generation efficient therapy for T1DM may help to improve the quality of life of diabetic patients who routinely employ insulin by the subcutaneous route. In this paper, we have depicted various next generation nanocarrier based insulin delivery systems such as chitosan-insulin nanoparticles, PLGA-insulin nanoparticles, dextran-insulin nanoparticles, polyalkylcyanoacrylated-insulin nanoparticles and solid lipid-insulin nanoparticles. Modulation of these insulin nanocarriers may lead to successful oral or pulmonary insulin nanoformulations in future clinical settings. Therefore, applications and limitations of these nanoparticles in delivering insulin to the targeted site have been thoroughly discussed.
Collapse
Affiliation(s)
- Garima Sharma
- Institute For Skeletal Aging, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 200704, Korea. .,Amity Institute of Nanotechnology, Amity University, Noida, Uttar Pradesh, India.
| | - Ashish Ranjan Sharma
- Institute For Skeletal Aging, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 200704, Korea.
| | - Ju-Suk Nam
- Institute For Skeletal Aging, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 200704, Korea.
| | - George Priya C Doss
- Medical Biotechnology Division, School of Biosciences and Technology, VIT University, Vellore, 632014, Tamil Nadu, India.
| | - Sang-Soo Lee
- Institute For Skeletal Aging, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 200704, Korea.
| | - Chiranjib Chakraborty
- Institute For Skeletal Aging, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 200704, Korea. .,Department of Bio-informatics, School of Computer and Information Sciences, Galgotias University, Greater Noida, India.
| |
Collapse
|
46
|
Ansari M. Oral Delivery of Insulin for Treatment of Diabetes: Classical Challenges and Current Opportunities. JOURNAL OF MEDICAL SCIENCES 2015. [DOI: 10.3923/jms.2015.209.220] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
47
|
Fox CB, Kim J, Le LV, Nemeth CL, Chirra HD, Desai TA. Micro/nanofabricated platforms for oral drug delivery. J Control Release 2015; 219:431-444. [PMID: 26244713 DOI: 10.1016/j.jconrel.2015.07.033] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/29/2015] [Accepted: 07/30/2015] [Indexed: 12/18/2022]
Abstract
The oral route of drug administration is most preferred due to its ease of use, low cost, and high patient compliance. However, the oral uptake of many small molecule drugs and biotherapeutics is limited by various physiological barriers, and, as a result, drugs suffer from issues with low solubility, low permeability, and degradation following oral administration. The flexibility of micro- and nanofabrication techniques has been used to create drug delivery platforms designed to address these barriers to oral drug uptake. Specifically, micro/nanofabricated devices have been designed with planar, asymmetric geometries to promote device adhesion and unidirectional drug release toward epithelial tissue, thereby prolonging drug exposure and increasing drug permeation. Furthermore, surface functionalization, nanotopography, responsive drug release, motion-based responses, and permeation enhancers have been incorporated into such platforms to further enhance drug uptake. This review will outline the application of micro/nanotechnology to specifically address the physiological barriers to oral drug delivery and highlight technologies that may be incorporated into these oral drug delivery systems to further enhance drug uptake.
Collapse
Affiliation(s)
- Cade B Fox
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Jean Kim
- UC Berkeley & UCSF Graduate Program in Bioengineering, UCSF Mission Bay Campus, San Francisco, CA 94158, USA
| | - Long V Le
- UC Berkeley & UCSF Graduate Program in Bioengineering, UCSF Mission Bay Campus, San Francisco, CA 94158, USA
| | - Cameron L Nemeth
- UC Berkeley & UCSF Graduate Program in Bioengineering, UCSF Mission Bay Campus, San Francisco, CA 94158, USA
| | - Hariharasudhan D Chirra
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA; UC Berkeley & UCSF Graduate Program in Bioengineering, UCSF Mission Bay Campus, San Francisco, CA 94158, USA.
| |
Collapse
|
48
|
Welling SH, Clemmensen LKH, Buckley ST, Hovgaard L, Brockhoff PB, Refsgaard HHF. In silico modelling of permeation enhancement potency in Caco-2 monolayers based on molecular descriptors and random forest. Eur J Pharm Biopharm 2015; 94:152-9. [PMID: 26004819 DOI: 10.1016/j.ejpb.2015.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/14/2015] [Accepted: 05/17/2015] [Indexed: 10/23/2022]
Abstract
Structural traits of permeation enhancers are important determinants of their capacity to promote enhanced drug absorption. Therefore, in order to obtain a better understanding of structure-activity relationships for permeation enhancers, a Quantitative Structural Activity Relationship (QSAR) model has been developed. The random forest-QSAR model was based upon Caco-2 data for 41 surfactant-like permeation enhancers from Whitehead et al. (2008) and molecular descriptors calculated from their structure. The QSAR model was validated by two test-sets: (i) an eleven compound experimental set with Caco-2 data and (ii) nine compounds with Caco-2 data from literature. Feature contributions, a recent developed diagnostic tool, was applied to elucidate the contribution of individual molecular descriptors to the predicted potency. Feature contributions provided easy interpretable suggestions of important structural properties for potent permeation enhancers such as segregation of hydrophilic and lipophilic domains. Focusing on surfactant-like properties, it is possible to model the potency of the complex pharmaceutical excipients, permeation enhancers. For the first time, a QSAR model has been developed for permeation enhancement. The model is a valuable in silico approach for both screening of new permeation enhancers and physicochemical optimisation of surfactant enhancer systems.
Collapse
Affiliation(s)
- Søren H Welling
- Global Research, Novo Nordisk A/S, Novo Nordisk Park, 2760 Måløv, Denmark; Technical University of Denmark, DTU Compute, 2800 Kgs. Lyngby, Denmark
| | | | - Stephen T Buckley
- Global Research, Novo Nordisk A/S, Novo Nordisk Park, 2760 Måløv, Denmark
| | - Lars Hovgaard
- Global Research, Novo Nordisk A/S, Novo Nordisk Park, 2760 Måløv, Denmark
| | - Per B Brockhoff
- Technical University of Denmark, DTU Compute, 2800 Kgs. Lyngby, Denmark
| | | |
Collapse
|
49
|
Karavasili C, Spanakis M, Papagiannopoulou D, Vizirianakis IS, Fatouros DG, Koutsopoulos S. Bioactive self-assembling lipid-like peptides as permeation enhancers for oral drug delivery. J Pharm Sci 2015; 104:2304-11. [PMID: 25994901 DOI: 10.1002/jps.24484] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 04/12/2015] [Accepted: 04/16/2015] [Indexed: 01/28/2023]
Abstract
Amphiphilic, lipid-like, self-assembling peptides are functional biomaterials with surfactant properties. In this work, lipid-like peptides were designed to have a hydrophilic head composed of aspartic acid or lysine and a six alanine residue hydrophobic domain and have a length similar to that of biological lipids. The aim of this work was to examine the potential of using ac-A6 K-CONH2 , KA6 -CONH2 , ac-A6 D-COOH, and DA6 -COOH lipid-like peptides as permeability enhancers to facilitate transport through the intestinal barrier. In vitro transport studies of the macromolecular fluorescent marker fluorescein isothiocyanate (FITC)-dextran (4.4 kDa) through Caco-2 cell monolayers show the permeation enhancement ability of the lipid-like peptides. We observed increased FITC-dextran transport across the epithelial monolayer up to 7.6-fold in the presence of lipid-like peptides. Furthermore, we monitored the transepithelial resistance and performed immunofluorescence studies of the cell tight junctions. Ex vivo studies showed increased mucosal to serosal absorption of FITC-dextran in rat jejunum in the presence of the ac-A6 D-COOH peptide. Furthermore, a small increase in the serosal transport of bovine serum albumin was observed upon addition of ac-A6 D-COOH. Lipid-like peptides are biocompatible and they do not affect epithelial cell viability and epithelial monolayer integrity. Our results suggest that short, lipid-like peptides may be used as permeation enhancers to facilitate oral delivery of diagnostic and therapeutic molecules.
Collapse
Affiliation(s)
- Christina Karavasili
- School of Pharmacy, Aristotle University of Thessaloniki, Department of Pharmaceutical Technology, Thessaloniki, GR-54124, Greece
| | - Marios Spanakis
- School of Pharmacy, Aristotle University of Thessaloniki, Department of Pharmacology and Molecular Biology, Thessaloniki, GR-54124, Greece
| | - Dionysia Papagiannopoulou
- School of Pharmacy, Aristotle University of Thessaloniki, Department of Pharmaceutical Chemistry, Thessaloniki, GR-54124, Greece
| | - Ioannis S Vizirianakis
- School of Pharmacy, Aristotle University of Thessaloniki, Department of Pharmacology and Molecular Biology, Thessaloniki, GR-54124, Greece
| | - Dimitrios G Fatouros
- School of Pharmacy, Aristotle University of Thessaloniki, Department of Pharmaceutical Technology, Thessaloniki, GR-54124, Greece
| | - Sotirios Koutsopoulos
- Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| |
Collapse
|
50
|
Song KH, Kim SB, Shim CK, Chung SJ, Kim DD, Rhee SK, Choi GJ, Kim CH, Kim K. Paracellular permeation-enhancing effect of AT1002 C-terminal amidation in nasal delivery. Drug Des Devel Ther 2015; 9:1815-23. [PMID: 25848218 PMCID: PMC4383222 DOI: 10.2147/dddt.s79383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The identification of permeation enhancers has gained interest in the development of drug delivery systems. A six-mer peptide, H-FCIGRL-OH (AT1002), is a tight junction modulator with promising permeation-enhancing activity. AT1002 enhances the transport of molecular weight markers or agents with low bioavailability with no cytotoxicity. However, AT1002 is not stable in neutral pH or after incubation under physiological conditions, which is necessary to fully uncover its permeation-enhancing effect. Thus, we increased the stability or mitigated the instability of AT1002 by modifying its terminal amino acids and evaluated its subsequent biological activity. METHODS C-terminal-amidated (FCIGRL-NH2, Pep1) and N-terminal-acetylated (Ac-FCIGRL, Pep2) peptides were analyzed by liquid chromatography-mass spectrometry. We further assessed cytotoxicity on cell monolayers, as well as the permeation-enhancing activity following nasal administration of the paracellular marker mannitol. RESULTS Pep1 was nontoxic to cell monolayers and showed a relatively low decrease in peak area compared to AT1002. In addition, administration of mannitol with Pep1 resulted in significant increases in the area under the plasma concentration-time curve and peak plasma concentration at 3.63-fold and 2.68-fold, respectively, compared to mannitol alone. In contrast, no increase in mannitol concentration was shown with mannitol/AT1002 or mannitol/Pep2 compared to the control. Thus, Pep1 increased the stability or possibly reduced the instability of AT1002, which resulted in an increased permeation-enhancing effect of AT1002. CONCLUSION These results suggest the potential usefulness of C-terminal-amidated AT1002 in enhancing nasal drug delivery, which may lead to the development of a practical drug delivery technology for drugs with low bioavailability.
Collapse
Affiliation(s)
- Keon-Hyoung Song
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan, Republic of Korea
| | - Sang-Bum Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Chang-Koo Shim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Suk-Jae Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Dae-Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang-Ki Rhee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan, Republic of Korea
| | - Guang J Choi
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan, Republic of Korea
| | - Chul-Hyun Kim
- Department of Sports Medicine, Soonchunhyang University, Asan, Republic of Korea
| | - Kiyoung Kim
- Department of Medical Biotechnology, Soonchunhyang University, Asan, Republic of Korea
| |
Collapse
|