1
|
Suri K, Pfeifer L, Cvet D, Li A, McCoy M, Singh A, Amiji MM. Oral delivery of stabilized lipid nanoparticles for nucleic acid therapeutics. Drug Deliv Transl Res 2024:10.1007/s13346-024-01709-4. [PMID: 39320435 DOI: 10.1007/s13346-024-01709-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Gastrointestinal disorders originate in the gastrointestinal tract (GIT), and the therapies can benefit from direct access to the GIT achievable through the oral route. RNA molecules show great promise therapeutically but are highly susceptible to degradation and often require a carrier for cytoplasmic access. Lipid nanoparticles (LNPs) are clinically proven drug-delivery agents, primarily administered parenterally. An ideal Orally Delivered (OrD) LNP formulation should overcome the diverse GI environment, successfully delivering the drug to the site of action. A versatile OrD LNP formulation has been developed to encapsulate and deliver siRNA and mRNA in this paper. The formulations were prepared by the systematic addition of cationic lipid to the base LNP formulation, keeping the total of cationic lipid and ionizable lipid to 50 mol%. Biorelevant media stability depicted increased resistance to bile salt mediated destabilization upon the addition of the cationic lipid, however the in vitro efficacy data underscored the importance of the ionizable lipid. Based on this, OrD LNP was selected comprising of 20% cationic lipid and 30% ionizable lipid. Further investigation revealed the enhanced efficacy of OrD LNP in vitro after incubation in different dilutions of fasted gastric, fasted intestinal media, and mucin. Confocal imaging and flow cytometry confirmed uptake while in vivo studies demonstrated efficacy with siRNA and mRNA as payloads. Taken together, this research introduces OrD LNP to deliver nucleic acid locally to the GIT.
Collapse
Affiliation(s)
- Kanika Suri
- Takeda Development Center Americas, Cambridge, MA, USA
- Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA, USA
| | - Liam Pfeifer
- Takeda Development Center Americas, Cambridge, MA, USA
| | - Donna Cvet
- Takeda Development Center Americas, Cambridge, MA, USA
| | - Angela Li
- Takeda Development Center Americas, Cambridge, MA, USA
| | - Michael McCoy
- Takeda Development Center Americas, Cambridge, MA, USA
| | - Amit Singh
- Takeda Development Center Americas, Cambridge, MA, USA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, Bouve College of Health Sciences, Northeastern University, Boston, MA, USA.
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
2
|
Morris G, Goodman S, Sorzabal Bellido I, Milanese C, Girella A, Pallavicini P, Taglietti A, Gaboardi M, Jäckel F, Diaz Fernandez YA, Raval R. Temperature and pH Stimuli-Responsive System Delivers Location-Specific Antimicrobial Activity with Natural Products. ACS APPLIED BIO MATERIALS 2024; 7:131-143. [PMID: 38079569 PMCID: PMC10792665 DOI: 10.1021/acsabm.3c00588] [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: 07/26/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 01/16/2024]
Abstract
Smart materials with controlled stimuli-responsive functions are at the forefront of technological development. In this work, we present a generic strategy that combines simple components, physicochemical responses, and easy fabrication methods to achieve a dual stimuli-responsive system capable of location-specific antimicrobial cargo delivery. The encapsulated system is fabricated by combining a biocompatible inert polymeric matrix of poly(dimethylsiloxane) (PDMS) and a bioactive cargo of saturated fatty acids. We demonstrate the effectiveness of our approach to deliver antimicrobial activity for the model bacteria Escherichia coli. The system responds to two control variables, temperature and pH, delivering two levels of antimicrobial response under distinct combinations of stimuli: one response toward the planktonic media and another response directly at the surface for sessile bacteria. Spatially resolved Raman spectroscopy alongside thermal and structural material analysis reveals that the system not only exhibits ON/OFF states but can also control relocation and targeting of the active cargo toward either the surface or the liquid media, leading to different ON/OFF states for the planktonic and sessile bacteria. The approach proposed herein is technologically simple and scalable, facing low regulatory barriers within the food and healthcare sectors by using approved components and relying on fundamental chemical processes. Our results also provide a proof-of-concept platform for the design and easy fabrication of delivery systems capable of operating as Boolean logic gates, delivering different responses under different environmental conditions.
Collapse
Affiliation(s)
- Gareth Morris
- Open
Innovation Hub for Antimicrobial Surfaces, Surface Science Research
Centre, University of Liverpool, Liverpool L69 3BX, U.K.
- Department
of Physics and Stephenson Institute for Renewable Energy, University of Liverpool, Liverpool L69 7ZE, U.K.
| | - Sean Goodman
- Open
Innovation Hub for Antimicrobial Surfaces, Surface Science Research
Centre, University of Liverpool, Liverpool L69 3BX, U.K.
| | - Ioritz Sorzabal Bellido
- Open
Innovation Hub for Antimicrobial Surfaces, Surface Science Research
Centre, University of Liverpool, Liverpool L69 3BX, U.K.
| | - Chiara Milanese
- Department
of Chemistry, University of Pavia, Via Taramelli 12, Pavia 27100, Italy
| | - Alessandro Girella
- Department
of Chemistry, University of Pavia, Via Taramelli 12, Pavia 27100, Italy
| | | | - Angelo Taglietti
- Department
of Chemistry, University of Pavia, Via Taramelli 12, Pavia 27100, Italy
| | - Mattia Gaboardi
- Materials
Physics Center, CSIC-UPV/EHU, Donostia - San Sebastian 20018, Spain
| | - Frank Jäckel
- Department
of Physics and Stephenson Institute for Renewable Energy, University of Liverpool, Liverpool L69 7ZE, U.K.
| | - Yuri A. Diaz Fernandez
- Open
Innovation Hub for Antimicrobial Surfaces, Surface Science Research
Centre, University of Liverpool, Liverpool L69 3BX, U.K.
- Department
of Chemistry, University of Pavia, Via Taramelli 12, Pavia 27100, Italy
| | - Rasmita Raval
- Open
Innovation Hub for Antimicrobial Surfaces, Surface Science Research
Centre, University of Liverpool, Liverpool L69 3BX, U.K.
| |
Collapse
|
3
|
Yosef AM, Alqarni RS, Sayd FY, Alhawiti MS, Almahlawi RM, Prabahar K, Uthumansha U, Alanazi MA, El-Sherbiny M, Elsherbiny N, Qushawy M. Preparation and Characterization of Novel Polyelectrolyte Liposomes Using Chitosan Succinate Layered over Chitosomes: A Potential Strategy for Colon Cancer Treatment. Biomedicines 2024; 12:126. [PMID: 38255231 PMCID: PMC10813275 DOI: 10.3390/biomedicines12010126] [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: 12/01/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
Chitosan succinate is distinguished by its ability to shield the loaded drug from the acidic environment, localize and keep the drug at the colon site, and release the drug over an extended time at basic pH. The current study attempts to develop polyelectrolyte liposomes (PEL), using chitosan and chitosan succinate (CSSC), as a carrier for liposomal-assisted colon target delivery of 5 fluorouracil (5FU). The central composite design was used to obtain an optimized formulation of 5FU-chitosomes. The chitosan-coated liposomes (chitosomes) were prepared by thin lipid film hydration technique. After that, the optimized formulation was coated with CSSC, which has several carboxylic (COOH) groups that produce an anionic charge that interacts with the cation NH2 in chitosan. The prepared 5FU-chitosomes formulations were evaluated for entrapment efficiency % (EE%), particle size, and in vitro drug release. The optimized 5FU-chitosomes formulation was examined for particle size, zeta potential, in vitro release, and mucoadhesive properties in comparison with the equivalent 5FU-liposomes and 5FU-PEL. The prepared 5FU-chitosomes exhibited high EE%, small particle size, low polydispersity index, and prolonged drug release. PEL significantly limited the drug release at acidic pH due to the deprotonation of carboxylate ions in CSSC, which resulted in strong repulsive forces, significant swelling, and prolonged drug release. According to a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay, PEL treatment significantly decreased the viability of HT-29 cells. When compared to 5FU-liposome and 5FU-chitosome, the in vivo pharmacokinetics characteristics of 5FU-PEL significantly (p < 0.05) improved. The findings show that PEL enhances 5FU permeability, which permits high drug concentrations to enter cells and inhibits the growth of colon cancer cells. Based on the current research, PEL may be used as a liposomal-assisted colon-specific delivery.
Collapse
Affiliation(s)
- Asmaa Mokhtar Yosef
- Pharm. D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.M.Y.); (R.S.A.); (F.Y.S.); (M.S.A.); (R.M.A.)
| | - Raghad Saleh Alqarni
- Pharm. D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.M.Y.); (R.S.A.); (F.Y.S.); (M.S.A.); (R.M.A.)
| | - Fai Yahya Sayd
- Pharm. D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.M.Y.); (R.S.A.); (F.Y.S.); (M.S.A.); (R.M.A.)
| | - Manar Saleem Alhawiti
- Pharm. D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.M.Y.); (R.S.A.); (F.Y.S.); (M.S.A.); (R.M.A.)
| | - Raghad M. Almahlawi
- Pharm. D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.M.Y.); (R.S.A.); (F.Y.S.); (M.S.A.); (R.M.A.)
| | - Kousalya Prabahar
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Ubaidulla Uthumansha
- Department of Pharmaceutics, Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai 600048, India;
| | - Mansuor A. Alanazi
- Department of Family and Community Medicine, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 13713, Saudi Arabia;
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Nehal Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Mona Qushawy
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
- Department of Pharmaceutics, Faculty of Pharmacy, Sinai University, Alarish 45511, North Sinai, Egypt
| |
Collapse
|
4
|
Chen M, Lan H, Jin K, Chen Y. Responsive nanosystems for targeted therapy of ulcerative colitis: Current practices and future perspectives. Drug Deliv 2023; 30:2219427. [PMID: 37288799 PMCID: PMC10405869 DOI: 10.1080/10717544.2023.2219427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/15/2023] [Accepted: 05/20/2023] [Indexed: 06/09/2023] Open
Abstract
The pharmacological approach to treating gastrointestinal diseases is suffering from various challenges. Among such gastrointestinal diseases, ulcerative colitis manifests inflammation at the colon site specifically. Patients suffering from ulcerative colitis notably exhibit thin mucus layers that offer increased permeability for the attacking pathogens. In the majority of ulcerative colitis patients, the conventional treatment options fail in controlling the symptoms of the disease leading to distressing effects on the quality of life. Such a scenario is due to the failure of conventional therapies to target the loaded moiety into specific diseased sites in the colon. Targeted carriers are needed to address this issue and enhance the drug effects. Conventional nanocarriers are mostly readily cleared and have nonspecific targeting. To accumulate the desired concentration of the therapeutic candidates at the inflamed area of the colon, smart nanomaterials with responsive nature have been explored recently that include pH responsive, reactive oxygen species responsive (ROS), enzyme responsive and thermo - responsive smart nanocarrier systems. The formulation of such responsive smart nanocarriers from nanotechnology scaffolds has resulted in the selective release of therapeutic drugs, avoiding systemic absorption and limiting the undesired delivery of targeting drugs into healthy tissues. Recent advancements in the field of responsive nanocarrier systems have resulted in the fabrication of multi-responsive systems i.e. dual responsive nanocarriers and derivitization that has increased the biological tissues and smart nanocarrier's interaction. In addition, it has also led to efficient targeting and significant cellular uptake of the therapeutic moieties. Herein, we have highlighted the latest status of the responsive nanocarrier drug delivery system, its applications for on-demand delivery of drug candidates for ulcerative colitis, and the prospects are underpinned.
Collapse
Affiliation(s)
- Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yun Chen
- Department of Colorectal Surgery, Xinchang People’s Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang, China
| |
Collapse
|
5
|
Preparation and Characterization of a Novel Multiparticulate Dosage Form Carrying Budesonide-Loaded Chitosan Nanoparticles to Enhance the Efficiency of Pellets in the Colon. Pharmaceutics 2022; 15:pharmaceutics15010069. [PMID: 36678698 PMCID: PMC9865799 DOI: 10.3390/pharmaceutics15010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
An attempt was made to conquer the limitation of orally administered nanoparticles for the delivery of budesonide to the colon. The ionic gelation technique was used to load budesonide on chitosan nanoparticles. The nanoparticles were investigated in terms of size, zeta potential, encapsulation efficiency, shape and drug release. Then, nanoparticles were pelletized using the extrusion-spheronization method and were investigated for their size, mechanical properties, and drug release. Pellets were subsequently coated with a polymeric solution composed of two enteric (eudragit L and S) and time-dependent polymers (eudragit RS) for colon-specific delivery. All formulations were examined for their anti-inflammatory effect in rats with induced colitis and the relapse of the colitis after discontinuation of treatment was also followed. The size of nanoparticles ranged between 288 ± 7.5 and 566 ± 7.7 nm and zeta potential verified their positive charged surface. The drug release from nanoparticles showed an initial burst release followed by a continuous release. Pelletized nanoparticles showed proper mechanical properties and faster drug release in acidic pH compared with alkaline pH. It was interesting to note that pelletized budesonide nanoparticles released the drug throughout the GIT in a sustained fashion, and had long-lasting anti-inflammatory effects while rapid relapse was observed for those treated with conventional budesonide pellets. It seems that there is a synergistic effect of nanoformulation of budesonide and the encapsulation of pelletized nanoparticles in a proper coating system for colon delivery that could result in a significant and long-lasting anti-inflammatory effect.
Collapse
|
6
|
Alghurabi H, Tagami T, Ogawa K, Ozeki T. Preparation, Characterization and In Vitro Evaluation of Eudragit S100-Coated Bile Salt-Containing Liposomes for Oral Colonic Delivery of Budesonide. Polymers (Basel) 2022; 14:2693. [PMID: 35808738 PMCID: PMC9268925 DOI: 10.3390/polym14132693] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to prepare a liposomal formulation of a model drug (budesonide) for colonic delivery by incorporating a bile salt (sodium glycocholate, SGC) into liposomes followed by coating with a pH-responsive polymer (Eudragit S100, ES100). The role of the SGC is to protect the liposome from the emulsifying effect of physiological bile salts, while that of ES100 is to protect the liposomes from regions of high acidity and enzymatic activity in the stomach and small intestine. Vesicles containing SGC were prepared by two preparation methods (sonication and extrusion), and then coated by ES100 (ES100-SGC-Lip). ES100-SGC-Lip showed a high entrapment efficiency (>90%) and a narrow size distribution (particle size = 275 nm, polydispersity index < 0.130). The characteristics of liposomes were highly influenced by the concentration of incorporated SGC. The lipid/polymer weight ratio, liposome charge, liposome addition, and mixing rate were critical factors for efficient and uniform coating. In vitro drug release studies in various simulated fluids indicate a pH-dependent dissolution of the coating layer, and the disintegration process of ES100-SGC-Lip was evaluated. In conclusion, the bile salt-containing ES100-coated liposomal formulation has potential for effective oral colonic drug delivery.
Collapse
Affiliation(s)
- Hamid Alghurabi
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (H.A.); (T.T.); (K.O.)
- Department of Pharmaceutics, College of Pharmacy, University of Kerbala, Kerbala 56001, Iraq
| | - Tatsuaki Tagami
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (H.A.); (T.T.); (K.O.)
| | - Koki Ogawa
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (H.A.); (T.T.); (K.O.)
| | - Tetsuya Ozeki
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (H.A.); (T.T.); (K.O.)
| |
Collapse
|
7
|
Sukocheva OA, Liu J, Neganova ME, Beeraka NM, Aleksandrova YR, Manogaran P, Grigorevskikh EM, Chubarev VN, Fan R. Perspectives of using microRNA-loaded nanocarriers for epigenetic reprogramming of drug resistant colorectal cancers. Semin Cancer Biol 2022; 86:358-375. [PMID: 35623562 DOI: 10.1016/j.semcancer.2022.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/07/2023]
Abstract
Epigenetic regulation by microRNAs (miRs) demonstrated a promising therapeutic potential of these molecules to regulate genetic activity in different cancers, including colorectal cancers (CRCs). The RNA-based therapy does not change genetic codes in tumor cells but can silence oncogenes and/or reactivate inhibited tumor suppressor genes. In many cancers, specific miRs were shown to promote or stop tumor progression. Among confirmed and powerful epigenetic regulators of colon carcinogenesis and development of resistance are onco-miRs, which include let-7, miR-21, miR-22, miR-23a, miR-27a, miR-34, miR-92, miR-96, miR-125b, miR-135b, miR-182, miR-200c, miR-203, miR-221, miR-421, miR-451, and others. Moreover, various tumor-suppressor miRs (miR-15b-5b, miR-18a, miR-20b, miR-22, miR-96, miR-139-5p, miR-145, miR-149, miR-197, miR-199b, miR-203, miR-214, miR-218, miR-320, miR-375-3p, miR-409-3p, miR-450b-5p, miR-494, miR-577, miR-874, and others) were found silenced in drug-resistant CRCs. Re-expression of tumor suppressor miR is complicated by the chemical nature of miRs that are not long-lasting compounds and require protection from the enzymatic degradation. Several recent studies explored application of miRs using nanocarrier complexes. This study critically describes the most successfully tested nanoparticle complexes used for intracellular delivery of nuclear acids and miRs, including micelles, liposomes, inorganic and polymeric NPs, dendrimers, and aptamers. Nanocarriers shield incorporated miRs and improve the agent stability in circulation. Attachment of antibodies and/or specific peptide or ligands facilitates cell-targeted miR delivery. Addressing in vivo challenges, a broad spectrum of non-toxic materials has been tested and indicated reliable advantages of lipid-based (lipoplexes) and polymer-based liposomes. Recent cutting-edge developments indicated that lipid-based complexes with multiple cargo, including several miRs, are the most effective approach to eradicate drug-resistant tumors. Focusing on CRC-specific miRs, this review provides a guidance and insights towards the most promising direction to achieve dramatic reduction in tumor growth and metastasis using miR-nanocarrier complexes.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou, 450052, China; The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Queensland, Australia; Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia.
| | - Junqi Liu
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou, 450052, China
| | - Margarita E Neganova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka, 142432, Russia
| | - Narasimha M Beeraka
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia; Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow, 119991, Russia; Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education and Research (JSS AHER), JSS Medical College, Mysuru, Karnataka, India
| | - Yulia R Aleksandrova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka, 142432, Russia
| | - Prasath Manogaran
- Translational Research Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Ekaterina M Grigorevskikh
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow, 119991, Russia
| | - Vladimir N Chubarev
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow, 119991, Russia
| | - Ruitai Fan
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou, 450052, China.
| |
Collapse
|
8
|
Wang G, Yang Y, Yi D, Yuan L, Yin PH, Ke X, Jun-Jie W, Tao MF. Eudragit S100 prepared pH-responsive liposomes-loaded betulinic acid against colorectal cancer in vitro and in vivo. J Liposome Res 2021; 32:250-264. [PMID: 34895013 DOI: 10.1080/08982104.2021.1999974] [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: 01/03/2023]
Abstract
This study aimed to develop polymer Eudragit S100 for preparing pH-responsive liposomes-loaded betulinic acid (pH-BA-LP) to improve the therapeutic index of chemotherapy for colorectal cancer. BA-loaded liposomes were coated with Eudragit S100 by a thin film dispersion and easily scalable pH-driven method. The prepared liposomes were evaluated for size, surface morphology, entrapment efficiency, stability, in vitro drug release, and antitumor activity. In particular, pH-BA-LP showed advantages such as lower size (<100 nm), encapsulation efficiency of 90%, high stability, and stably cumulative release. By detecting the antitumor effects of pH-BA-LP in vivo, it showed that the tumor proliferation and cell migration were significantly inhibited in colorectal cancer. The pH-BA-LP also inhibited tumor growth via the regulation of Akt/TLR-mediated signalling and significantly down-regulated the expression of NFAT1 and NFAT4 proteins. It was found that pH-BA-LP can increase NK cells and CD3+ cells in tumor tissues, and the proportion of CD8+ cells in CD3+ cells was also increased, which proved that pH-BA-LP can play an antitumor effect by enhancing the autoimmunity level in tumor-bearing mice. The positive infiltration rates of CD8 and CD68 were increased and CD163 was relatively decreased by using pH-BA-LP, which proved that pH-BA-LP can regulate the immune infiltration levels in tumor-bearing mice. Therefore, the present work provides an effective method to prepare pH-responsive polymer-coated liposomes for colonic delivery with biologically active compounds.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Yu Yang
- Jiangsu University School of Pharmacy, Zhenjiang City, China
| | - Du Yi
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Lu Yuan
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Pei-Hao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu Ke
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Jun-Jie
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Min-Fang Tao
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| |
Collapse
|
9
|
Panova IG, Sudareva EA, Novoskoltseva OA, Spiridonov VV, Shtilman MI, Richtering W, Yaroslavov AA. Temperature-induced unloading of liposomes bound to microgels. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
10
|
Yu C, Lu P, Liu S, Li Q, Xu E, Gong J, Liu S, Yang C. Efficiency of Deoxynivalenol Detoxification by Microencapsulated Sodium Metabisulfite Assessed via an In Vitro Bioassay Based on Intestinal Porcine Epithelial Cells. ACS OMEGA 2021; 6:8382-8393. [PMID: 33817499 PMCID: PMC8015119 DOI: 10.1021/acsomega.1c00117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/10/2021] [Indexed: 05/11/2023]
Abstract
Deoxynivalenol (DON) contamination occurs in feeds and causes a reduction in growth performance, damage to the intestinal epithelial cells, and increased susceptibility to enteric pathogen challenge. Sodium metabisulfite (SMBS) has shown promise in reducing DON; however, SMBS quickly degrades under aqueous acidic conditions such as the environment within a stomach. Thus, protection of SMBS is required for effective delivery to the small intestine to detoxify DON. This study was to encapsulate SMBS into hydrogenated palm oil-based microparticles for its delivery to the small intestine and to evaluate its efficacy on DON detoxification in simulated intestinal fluids using IPEC-J2 cells in vitro. The diameter of the SMBS containing microparticles was 511 ± 135 μm, and the loading capacity of SMBS in the microparticles was 45.50%; 1.41% of the encapsulated SMBS (ES) was released into the simulated gastric fluid, and 66.39% of ES was progressively released into the simulated intestinal fluid within 4 h at 37 °C. In IPEC-J2 cells, when DON was treated with the simulated gastric fluid containing 0.5% ES for 2 h, then mixed with the simulated intestinal fluid (1:1) and incubated for 2 h, cytotoxicity was not observed. DON treated with 0.5 ES decreased the gene expression of inflammatory cytokines in the cells compared with DON alone and maintained the cell integrity. To conclude, the SMBS containing microparticles were stable in the simulated gastric fluid and allowed a progressive release of SMBS in the simulated intestinal fluid. The released SMBS in the simulated intestinal fluid effectively detoxified DON.
Collapse
Affiliation(s)
- Changning Yu
- Department
of Biosystems Engineering, University of
Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Peng Lu
- Department
of Animal Science, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Shangxi Liu
- Department
of Animal Science, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Qiao Li
- Department
of Animal Science, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Erhua Xu
- King
Techina Group, No. 8,
Yinxing Road, Renhe Street, Yuhang District, Hangzhou 311107, China
| | - Joshua Gong
- Guelph
Research and Development Centre, Agriculture Agri-Food Canada, Guelph, Ontario N1G 5C9, Canada
| | - Song Liu
- Department
of Biosystems Engineering, University of
Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Chengbo Yang
- Department
of Animal Science, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| |
Collapse
|
11
|
De Leo V, Milano F, Agostiano A, Catucci L. Recent Advancements in Polymer/Liposome Assembly for Drug Delivery: From Surface Modifications to Hybrid Vesicles. Polymers (Basel) 2021; 13:1027. [PMID: 33810273 PMCID: PMC8037206 DOI: 10.3390/polym13071027] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Liposomes are consolidated and attractive biomimetic nanocarriers widely used in the field of drug delivery. The structural versatility of liposomes has been exploited for the development of various carriers for the topical or systemic delivery of drugs and bioactive molecules, with the possibility of increasing their bioavailability and stability, and modulating and directing their release, while limiting the side effects at the same time. Nevertheless, first-generation vesicles suffer from some limitations including physical instability, short in vivo circulation lifetime, reduced payload, uncontrolled release properties, and low targeting abilities. Therefore, liposome preparation technology soon took advantage of the possibility of improving vesicle performance using both natural and synthetic polymers. Polymers can easily be synthesized in a controlled manner over a wide range of molecular weights and in a low dispersity range. Their properties are widely tunable and therefore allow the low chemical versatility typical of lipids to be overcome. Moreover, depending on their structure, polymers can be used to create a simple covering on the liposome surface or to intercalate in the phospholipid bilayer to give rise to real hybrid structures. This review illustrates the main strategies implemented in the field of polymer/liposome assembly for drug delivery, with a look at the most recent publications without neglecting basic concepts for a simple and complete understanding by the reader.
Collapse
Affiliation(s)
- Vincenzo De Leo
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy;
| | - Francesco Milano
- Istituto di Scienze delle Produzioni Alimentari (ISPA), Consiglio Nazionale delle Ricerche (CNR), S.P. Lecce-Monteroni, Ecotekne, 73100 Lecce, Italy;
| | - Angela Agostiano
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy;
| | - Lucia Catucci
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy;
| |
Collapse
|
12
|
Andretto V, Rosso A, Briançon S, Lollo G. Nanocomposite systems for precise oral delivery of drugs and biologics. Drug Deliv Transl Res 2021; 11:445-470. [PMID: 33534107 DOI: 10.1007/s13346-021-00905-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 12/15/2022]
Abstract
Oral delivery is considered the favoured route of administration for both local and systemic delivery of active molecules. Formulation of drugs in conventional systems and nanoparticles has provided opportunities for targeting the gastrointestinal (GI) tract, increasing drug solubility and bioavailability. Despite the achievements of these delivery approaches, the development of a product with the ability of delivering drug molecules at a specific site and according to patients' needs remains a challenging endeavour. The complexity of the physicochemical properties of colloidal systems, their stability in different regions of the gastrointestinal tract, and interaction with the restrictive biological barriers hampered their success for oral precise medicine. To overcome these issues, nanoparticles have been combined with polymers to create hybrid nanosystems, namely nanocomposites. They offer enormous possibilities of structural and mechanical modifications to both nanoparticles and polymeric matrixes to generate systems with new properties, functions, and applications for oral delivery. In this review, nanocomposites' physicochemical and functional properties intended to target specific regions of the GI tract-oral cavity, stomach, small bowel, and colon-are analysed. In parallel, it is provided an insight in the nanocomposite solutions for oral delivery intended for systemic and local absorption, together with a focus on inflammatory bowel diseases (IBDs). Additional difficulties in managing IBD related to the alteration in the physiology of the intestine are described. Finally, future perspectives and opportunities for advancement in this field are discussed.
Collapse
Affiliation(s)
- Valentina Andretto
- LAGEPP UMR 5007, Univ Lyon, Université Claude Bernard Lyon 1, CNRS, 43 Boulevard du 11 Novembre 1918, 69100, Villeurbanne, France
| | - Annalisa Rosso
- LAGEPP UMR 5007, Univ Lyon, Université Claude Bernard Lyon 1, CNRS, 43 Boulevard du 11 Novembre 1918, 69100, Villeurbanne, France
| | - Stéphanie Briançon
- LAGEPP UMR 5007, Univ Lyon, Université Claude Bernard Lyon 1, CNRS, 43 Boulevard du 11 Novembre 1918, 69100, Villeurbanne, France
| | - Giovanna Lollo
- LAGEPP UMR 5007, Univ Lyon, Université Claude Bernard Lyon 1, CNRS, 43 Boulevard du 11 Novembre 1918, 69100, Villeurbanne, France.
| |
Collapse
|
13
|
Román-Aguirre M, Leyva-Porras C, Cruz-Alcantar P, Aguilar-Elguézabal A, Saavedra-Leos MZ. Comparison of Polysaccharides as Coatings for Quercetin-Loaded Liposomes (QLL) and Their Effect as Antioxidants on Radical Scavenging Activity. Polymers (Basel) 2020; 12:polym12122793. [PMID: 33255914 PMCID: PMC7760579 DOI: 10.3390/polym12122793] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Liposomes are microstructures containing lipid and aqueous phases employed in the encapsulation and delivery of bioactive agents. Quercetin-loaded liposomes (QLLs) were coated with three different polysaccharides and then tested as radical scavengers. Lactose (LCQLL), chitosan (CCQLL), and inulin (ICQLL) were employed as coating materials. Particle size determined by light scattering, showed primary size of 200 nm for all samples, while a secondary particle size of 600 nm was observed for CCQLL. Scanning electron microscopy (SEM) evidenced particle aggregation with the addition of the polysaccharide coating. Transmission electron microscopy (TEM) revealed the layered microstructure of liposomes composed of at least two layers, and primary particle size below 100 nm. QLL showed higher antioxidant activity than the coated liposomes. This behavior was attributed to the chemical interaction between quercetin and the corresponding coating polysaccharide in the layered structure, which traps the quercetin and keeps it unavailable for radical scavenging. From the three polysaccharides, lactose showed a better performance as coating material in the antioxidant activity, which suggested that the smaller size of the disaccharide molecule resulted in a faster releasing of the quercetin in the solution. Thus, LCQLL is an advantageous way to deliver quercetin for antioxidant purposes, where the low stability in delivered media of quercetin loaded liposomes is commonly compromised.
Collapse
Affiliation(s)
- Manuel Román-Aguirre
- Doctorado Institucional en Ingeniería y Ciencia de los Materiales, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, Mexico;
| | - César Leyva-Porras
- Laboratorio Nacional de Nanotecnología (NanoTech), Centro de Investigación en Materiales Avanzados S.C. (CIMAV), Chihuahua 31136, Mexico;
| | - Pedro Cruz-Alcantar
- Coordinación Académica Región Altiplano (COARA), Universidad Autónoma de San Luis Potosí, Matehuala 78700, Mexico;
| | - Alfredo Aguilar-Elguézabal
- Departamento de Ingeniería y Química de Materiales, Centro de Investigación en Materiales Avanzados, S.C. (CIMAV), Chihuahua 31136, Mexico;
| | - María Zenaida Saavedra-Leos
- Coordinación Académica Región Altiplano (COARA), Universidad Autónoma de San Luis Potosí, Matehuala 78700, Mexico;
- Correspondence: ; Tel.: +52-(488)-1250150
| |
Collapse
|
14
|
Kim JM, Kim DH, Park HJ, Ma HW, Park IS, Son M, Ro SY, Hong S, Han HK, Lim SJ, Kim SW, Cheon JH. Nanocomposites-based targeted oral drug delivery systems with infliximab in a murine colitis model. J Nanobiotechnology 2020; 18:133. [PMID: 32933548 PMCID: PMC7493402 DOI: 10.1186/s12951-020-00693-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/09/2020] [Indexed: 02/08/2023] Open
Abstract
Background Infliximab (IFX), a TNF-α blocking chimeric monoclonal antibody, induces clinical response and mucosal healing in patients with inflammatory bowel disease (IBD). However, systemic administration of this agent causes unwanted side effects. Oral delivery of antibody therapeutics might be an effective treatment strategy for IBD compared to intravenous administration. Results All three carriers had a high encapsulation efficiency, narrow size distribution, and minimal systemic exposure. There was a higher interaction between nanocomposite carriers and monocytes compared to lymphocytes in the PBMC of IBD patients. Orally administered nanocomposite carriers targeted to inflamed colitis minimized systemic exposure. All IFX delivery formulations with nanocomposite carriers had a significantly less colitis-induced body weight loss, colon shortening and histomorphological score, compared to the DSS-treated group. AC-IFX-L and EAC-IFX-L groups showed significantly higher improvement of the disease activity index, compared to the DSS-treated group. In addition, AC-IFX-L and EAC-IFX-L alleviated pro-inflammatory cytokine expressions (Tnfa, Il1b, and Il17). Conclusion We present orally administered antibody delivery systems which improved efficacy in murine colitis while reducing systemic exposure. These oral delivery systems suggest a promising therapeutic approach for treating IBD.![]()
Collapse
Affiliation(s)
- Jung Min Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Da Hye Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Jeong Park
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea
| | - Hyun Woo Ma
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - I Seul Park
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Mijeong Son
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - So Youn Ro
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea
| | - Seokmann Hong
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea.,Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, 05006, Republic of Korea
| | - Hyo Kyung Han
- College of Pharmacy, Dongguk University-Seoul, Dongguk‑ro‑32, Ilsan‑donggu, Goyang, South Korea
| | - Soo Jeong Lim
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea
| | - Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea. .,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea. .,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
15
|
Goel H, Razdan K, Singla R, Talegaonkar S, Khurana RK, Tiwary AK, Sinha VR, Singh KK. Engineered Site-specific Vesicular Systems for Colonic Delivery: Trends and Implications. Curr Pharm Des 2020; 26:5441-5455. [PMID: 32787754 DOI: 10.2174/1381612826666200813132301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/25/2020] [Indexed: 12/13/2022]
Abstract
Steering drug-loaded, site-specific, coated lipid vesicles to the target receptor sites have the potential of plummeting adverse effects and improving the pharmacological response in diverse pathologies of the large bowel, especially the colon. Colonic delivery via oral route has its own challenges, often governed by several glitches such as drug degradation or absorption in the upper GIT, instability of proteins/peptides due to high molecular weight, and peptidase activity in the stomach. Consequently, colon-specific coated liposomal systems (CSLS) offer a potential alternate for not only site-specificity, but protection from proteolytic activity, and prolonged residence time for greater systemic bioavailability. On the other hand, liposomal delivery via the oral route is also cumbersome owing to several barriers such as instability in GIT, difficulty in crossing membranes, and issues related to production at the pilot scale. New advancements in the field of CSLS have successfully improved the stability and permeability of liposomes for oral delivery via modulating the compositions of lipid bilayers, adding polymers or ligands. Despite this ostensible propitiousness, no commercial oral CSLS has advanced from bench to bedside for targeted delivery to the colon as yet. Nevertheless, CSLS has quite fascinated the manufacturers owing to its potential industrial viability, simplistic and low-cost design. Hence, this review aims to decipher the convolutions involved in the engineering process of industrially viable CSLS for colonic delivery.
Collapse
Affiliation(s)
- Honey Goel
- University Institute of Pharmaceutical Sciences and Research, Baba Farid University of Health Sciences, Faridkot, India
| | - Karan Razdan
- School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston, United Kingdom
| | - Richu Singla
- Department of Microbiology, Viral Research Diagnostics Laboratory (VRDL), Guru Gobind Singh Medical College and Hospital, Baba Farid University of Health Sciences, Faridkot, India
| | | | - Rajneet Kaur Khurana
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Ashok Kumar Tiwary
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Vivek Ranjan Sinha
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Kamalinder K Singh
- School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston, United Kingdom
| |
Collapse
|
16
|
Taipaleenmäki E, Christensen G, Brodszkij E, Mouritzen SA, Gal N, Madsen S, Hedemann MS, Knudsen TA, Jensen HM, Christiansen SL, Sparsø FV, Städler B. Mucopenetrating polymer – Lipid hybrid nanovesicles as subunits in alginate beads as an oral formulation. J Control Release 2020; 322:470-485. [DOI: 10.1016/j.jconrel.2020.03.047] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/21/2020] [Accepted: 03/29/2020] [Indexed: 12/18/2022]
|
17
|
Jain SK, Jain AK, Rajpoot K. Expedition of Eudragit® Polymers in the Development of Novel Drug Delivery Systems. Curr Drug Deliv 2020; 17:448-469. [PMID: 32394836 DOI: 10.2174/1567201817666200512093639] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/10/2020] [Accepted: 04/20/2020] [Indexed: 12/16/2022]
Abstract
Eudragit® polymer has been widely used in film-coating for enhancing the quality of products over other materials (e.g., shellac or sugar). Eudragit® polymers are obtained synthetically from the esters of acrylic and methacrylic acid. For the last few years, they have shown immense potential in the formulations of conventional, pH-triggered, and novel drug delivery systems for incorporating a vast range of therapeutics including proteins, vitamins, hormones, vaccines, and genes. Different grades of Eudragit® have been used for designing and delivery of therapeutics at a specific site via the oral route, for instance, in stomach-specific delivery, intestinal delivery, colon-specific delivery, mucosal delivery. Further, these polymers have also shown their great aptitude in topical and ophthalmic delivery. Moreover, available literature evidences the promises of distinct Eudragit® polymers for efficient targeting of incorporated drugs to the site of interest. This review summarizes some potential researches that are being conducted by eminent scientists utilizing the distinct grades of Eudragit® polymers for efficient delivery of therapeutics at various sites of interest.
Collapse
Affiliation(s)
- Sunil Kumar Jain
- Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur (C.G.) 495 009, India
| | - Akhlesh K Jain
- Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur (C.G.) 495 009, India
| | - Kuldeep Rajpoot
- Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur (C.G.) 495 009, India
| |
Collapse
|
18
|
Halimi M, Alishahi M, Abbaspour MR, Ghorbanpoor M, Tabandeh MR. High efficacy and economical procedure of oral vaccination against Lactococcus garvieae/Streptococcus iniae in rainbow trout (Oncorhynchus mykiss). FISH & SHELLFISH IMMUNOLOGY 2020; 99:505-513. [PMID: 32092407 DOI: 10.1016/j.fsi.2020.02.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/10/2020] [Accepted: 02/16/2020] [Indexed: 06/10/2023]
Abstract
The present study was aimed to examine the efficacy of chitosan-alginate coated vaccines against pathogenicity of Lactococcus garvieae and Streptococcus iniae in rainbow trout. Fish were divided into four groups including: Group A: fish immunized by chitosan-alginate coated vaccine, Group B: fish immunized by non-coated vaccine, Group C: fish feed by chitosan-alginate coated pellets without vaccine and Group D: fish feed by basic diet (non-coated and without vaccine). In groups A and B, the vaccination was carried out for 14 days and after that supplemented with fundamental diet (control diet). Comparable to groups A and B, fish of group C were also fed 14 days with test diets and after that fed control food. On day 0, 20, 40 and 60 of the experiment, serum samples were given. Fish have been challenged with live L. garvieae and S. iniae after 60 days. The levels of bactericidal activity and complement activity among innate immunity components extended on day 20 of the research and after that decreased in group A and B (P < 0.05) all through the examination. The relative expression of IL-6 and IgM in groups A and B extended on examination day 20. The expression of these genes illustrated no advancements in different groups in during the examination (P > 0.05). In group A, the serum antibody titer against L. garvieae and S. iniae broadly raised on day 40 and 60 of examination, whereas in group B, the immune response titer against S. iniae and L. garvieae illustrated a significant elevation on day 60 of the trial (P < 0.05). After challenge with live bacteria, survival rate of 83 ± 9.1%(challenged with S. iniae) and 72.18 ± 9.8% (challenged with L. garvieae) were gotten independently in group A, which were higher than survival of other exploratory groups (P < 0.05). In conclusion, the results of the present examination appear that the orally vaccination of rainbow trout with chitosan-alginate covered vaccine stimulates immunity system and also efficiently protects rainbow trout against Lactococcus garvieae and Streptococcus iniae.
Collapse
Affiliation(s)
- Mostafa Halimi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Mojtaba Alishahi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohammad Reza Abbaspour
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoud Ghorbanpoor
- Department of Microbiology and Immunology,Faculty of Veterinary Medicine,Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohammad Reza Tabandeh
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
19
|
Jiménez-Avalos JA, Arrevillaga-Boni G, González-López L, García-Carvajal ZY, González-Avila M. Classical methods and perspectives for manipulating the human gut microbial ecosystem. Crit Rev Food Sci Nutr 2020; 61:234-258. [PMID: 32114770 DOI: 10.1080/10408398.2020.1724075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A healthy Human Gut Microbial Ecosystem (HGME) is a necessary condition for maintaining the orderly function of the whole body. Major alterations in the normal gut microbial composition, activity and functionality (dysbiosis) by an environmental or host-related disruptive event, can compromise metabolic, inflammatory, and neurological processes, causing disorders such as obesity, inflammatory bowel disease, colorectal cancer, and depressive episodes. The restore or the maintaining of the homeostatic balance of Gut Microbiota (GM) populations (eubiosis) is possible through diet, the use of probiotics, prebiotics, antibiotics, and even Fecal Microbiota Transplantation (FMT). Although these "classic methods" represent an effective and accepted way to modulate GM, the complexity of HGME requires new approaches to control it in a more appropriate way. Among the most promising emergent strategies for modulating GM are the use of engineered nanomaterials (metallic nanoparticles (NP), polymeric-NP, quantum dots, micelles, dendrimers, and liposomes); phagotherapy (i.e., phages linked with the CRISPR/Cas9 system), and the use of antimicrobial peptides, non-antibiotic drugs, vaccines, and immunoglobulins. Here we review the current state of development, implications, advantages, disadvantages, and perspectives of the different approaches for manipulating HGME.
Collapse
Affiliation(s)
- Jorge Armando Jiménez-Avalos
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | - Gerardo Arrevillaga-Boni
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | | | - Zaira Yunuen García-Carvajal
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | - Marisela González-Avila
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| |
Collapse
|
20
|
He H, Lu Y, Qi J, Zhu Q, Chen Z, Wu W. Adapting liposomes for oral drug delivery. Acta Pharm Sin B 2019; 9:36-48. [PMID: 30766776 PMCID: PMC6362257 DOI: 10.1016/j.apsb.2018.06.005] [Citation(s) in RCA: 349] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/21/2018] [Accepted: 04/12/2018] [Indexed: 02/08/2023] Open
Abstract
Liposomes mimic natural cell membranes and have long been investigated as drug carriers due to excellent entrapment capacity, biocompatibility and safety. Despite the success of parenteral liposomes, oral delivery of liposomes is impeded by various barriers such as instability in the gastrointestinal tract, difficulties in crossing biomembranes, and mass production problems. By modulating the compositions of the lipid bilayers and adding polymers or ligands, both the stability and permeability of liposomes can be greatly improved for oral drug delivery. This review provides an overview of the challenges and current approaches toward the oral delivery of liposomes.
Collapse
Key Words
- APC, antigen-presenting cell
- AUC, area under curve
- Absorption
- BSA, bovine serum albumin
- Bioavailability
- DC, dendritic cells
- DMPC, dimyristoyl phosphatidyl choline
- DPPC, dipalmitoyl phosphotidylcholine
- Drug delivery
- FAE, follicle-associated epithelia
- FITC, fluorescein isothiocyannate
- GIT, gastrointestinal tract
- LUV, large unilamellar vesicles
- Liposomes
- MLV, multilamellar vesicles
- MRT, mean residence time
- MVL, multivesicular liposomes
- Oral
- PC, phosphatidylcholine
- PEG, polyethylene glycol
- RES, reticulo-endothelial
- SC, sodium cholate
- SDC, sodium deoxycholate
- SGC, sodium glycocholate
- SPC, soy phosphatidylcholine
- STC, sodium taurocholate
- SUV, small unilamellar vesicles
- Stability
- TPGS, tocopherol polyethylene glycol succinate
- Tgel, gelling temperature
- Tp, phase transition temperature
- UEA 1, ulex europaeus agglutinin 1
- WGA, wheat germ agglutinin
- rhEGF, recombinant human epithelial growth factor
Collapse
Affiliation(s)
- Haisheng He
- Key Laboratory of Smart Drug Delivery of MOE and PLA, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi Lu
- Key Laboratory of Smart Drug Delivery of MOE and PLA, School of Pharmacy, Fudan University, Shanghai 201203, China
- Shanghai Dermatology Hospital, Shanghai 200443, China
| | - Jianping Qi
- Key Laboratory of Smart Drug Delivery of MOE and PLA, School of Pharmacy, Fudan University, Shanghai 201203, China
- Shanghai Dermatology Hospital, Shanghai 200443, China
| | - Quangang Zhu
- Shanghai Dermatology Hospital, Shanghai 200443, China
| | | | - Wei Wu
- Key Laboratory of Smart Drug Delivery of MOE and PLA, School of Pharmacy, Fudan University, Shanghai 201203, China
- Shanghai Dermatology Hospital, Shanghai 200443, China
| |
Collapse
|
21
|
Halimi M, Alishahi M, Abbaspour MR, Ghorbanpoor M, Tabandeh MR. Efficacy of a Eudragit L30D-55 encapsulated oral vaccine containing inactivated bacteria (Lactococcus garvieae/Streptococcus iniae) in rainbow trout (Oncorhynchus mykiss). FISH & SHELLFISH IMMUNOLOGY 2018; 81:430-437. [PMID: 30056210 DOI: 10.1016/j.fsi.2018.07.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 07/22/2018] [Accepted: 07/25/2018] [Indexed: 06/08/2023]
Abstract
The efficacy of a Eudragit L30D-55 encapsulated vaccine against Lactococcus garvieae and Streptococcus iniae was investigated in rainbow trout. Fish were divided into four groups and fed the different experimental feeds. Groups were: A) fish immunized by Eudragit-coated pellets containing vaccine, B) fish immunized by vaccine-coated pellets without Eudragit, C) fish fed Eudragit-coated pellets without vaccine and D) fish fed pellets without vaccine orEudragit (control group). In groups A and B, the vaccination was conducted for 14 days. Similar to groups A and B, fish of group C were fed 14 days with pellets coated with Eudragit and afterwards they were fed control diet. Serum samples were taken on day 0, 20, 40 and 60 of the experiment. After 60 days, fish were challenged with L. garvieae and S. iniae. In almost all groups, innate immunity components including alternative complement activity, lysozyme activity, bactericidal activity, IgM and total protein showed no significant changes during the 60 days that the experiment lasted. However, the blood respiratory burst activity and lysozyme activity showed a significant increase on day 20 of experiment in groups B and D respectively (P < 0.05). The relative expression of immune-related genes including IL-6 and IgM genes was higher in vaccinated fish, with the highest expression in those immunized by Eudragit-coated pellets (Group A). In addition, the relative expression of IL-6 and IgM peaked on day 20 but decreased on day 60 in vaccinated groups. The ELISA antibody titer against L. garvieae increased from day 20 and peaked on day 60 of experiment (P < 0.05). Also, the antibody titer against L. garvieae was higher in fish immunized by Eudragit-coated pellets (Group A) compared to fish of group C and control. After bacterial challenge, a survival percentages of % 85 ± 7.07% (challenged with S. iniae) and % 72.21 ± 7.8% (challenged with L. garvieae) were observed respectively in groups immunized with pellets coated with Eudragit L30D-55 (Group A), which were higher than survival percentages obtained in other experimental groups (P < 0.05). The results of the present study demonstrate that the oral administration of Eudragit L30D-55-encapsulated vaccine appropriately protects rainbow trout against Lactococcus garvieae and Streptococcus iniae.
Collapse
Affiliation(s)
- Mostafa Halimi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Mojtaba Alishahi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohammad Reza Abbaspour
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoud Ghorbanpoor
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohammad Reza Tabandeh
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
22
|
Preparation, characterization, and pharmacokinetics of liposomal docetaxel for oral administration. Arch Pharm Res 2018; 41:765-775. [PMID: 29961194 DOI: 10.1007/s12272-018-1046-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/11/2018] [Indexed: 12/19/2022]
Abstract
A docetaxel (DTX) liposomal formulation composed of egg phosphatidylcholine, sodium deoxycholate, and stearylamine was developed. Eudragit (0.5%) was coated to deliver the drug to the region between the distal small intestine and the colon. Lyophilized trehalose and mannitol were used as cryoprotectants because they preserve the particle integrity and good appearance. In vitro release studies showed that the amount of drug released from the coated liposomes was low in solution 1, which simulated the pH condition of the stomach. Especially during the average gastric emptying time, the amount of drug released decreased when Eudragit was added. The plasma DTX concentration was evaluated in pharmacokinetic studies. The plasma drug concentration after intravenous (i.v.) administration decreased rapidly within 120 min. Free DTX formulated using Tween 80 and the lyophilized Eudragit-coated liposomal formulation were compared after oral administration. The oral liposomal formulation had a longer half-life (t1/2) and three-fold higher oral bioavailability. Thus, lyophilized Eudragit-coated liposomal DTX could be a promising therapy for various solid tumors to improve patient convenience and quality of life.
Collapse
|
23
|
De Leo V, Milano F, Mancini E, Comparelli R, Giotta L, Nacci A, Longobardi F, Garbetta A, Agostiano A, Catucci L. Encapsulation of Curcumin-Loaded Liposomes for Colonic Drug Delivery in a pH-Responsive Polymer Cluster Using a pH-Driven and Organic Solvent-Free Process. Molecules 2018; 23:E739. [PMID: 29570636 PMCID: PMC6017095 DOI: 10.3390/molecules23040739] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/09/2018] [Accepted: 03/21/2018] [Indexed: 01/08/2023] Open
Abstract
The present study aimed to develop and optimize liposome formulation for the colonic delivery of biologically active compounds. A strategy to facilitate such targeting is to formulate liposomes with a polymer coating sensitive to the pH shifts in the gastrointestinal tract. To this end, liposomes encapsulating curcumin-chosen as the biologically active compound model-and coated with the pH-responsive polymer Eudragit S100 were prepared and characterized. Curcumin was encapsulated into small unilamellar vesicles (SUVs) by the micelle-to-vesicle transition method (MVT) in a simple and organic solvent-free way. Curcumin-loaded liposomes were coated with Eudragit S100 by a fast and easily scalable pH-driven method. The prepared liposomes were evaluated for size, surface morphology, entrapment efficiency, stability, in vitro drug release, and curcumin antioxidant activity. In particular, curcumin-loaded liposomes displayed size lower than 100 nm, encapsulation efficiency of 98%, high stability at both 4 °C and 25 °C, high in vitro antioxidant activity, and a cumulative release that was completed within 200 min. A good Eudragit S100 coating which did not alter the properties of the curcumin-loaded liposomes was obtained. The present work therefore provides a fast and solvent-free method to prepare pH-responsive polymer-coated liposomes for the colonic delivery of biologically active compounds.
Collapse
Affiliation(s)
- Vincenzo De Leo
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy.
- CNR-IPCF Institute for Physical and Chemical Processes, Bari unit, Via Orabona 4, 70126 Bari, Italy.
| | - Francesco Milano
- CNR-IPCF Institute for Physical and Chemical Processes, Bari unit, Via Orabona 4, 70126 Bari, Italy.
| | - Erminia Mancini
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy.
| | - Roberto Comparelli
- CNR-IPCF Institute for Physical and Chemical Processes, Bari unit, Via Orabona 4, 70126 Bari, Italy.
| | - Livia Giotta
- Department of Biological and Environmental Sciences and Technologies, University of Salento, SP Lecce-Monteroni, I-73100 Lecce, Italy.
| | - Angelo Nacci
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy.
- CNR-ICCOM Institute of chemistry of organometallic compounds, Bari unit, Via E. Orabona, 4, 70126 Bari, Italy.
| | | | - Antonella Garbetta
- CNR-ISPA Institute of Sciences of Food Production, Via G. Amendola 122/O, 70125 Bari, Italy.
| | - Angela Agostiano
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy.
- CNR-IPCF Institute for Physical and Chemical Processes, Bari unit, Via Orabona 4, 70126 Bari, Italy.
| | - Lucia Catucci
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy.
- CNR-IPCF Institute for Physical and Chemical Processes, Bari unit, Via Orabona 4, 70126 Bari, Italy.
| |
Collapse
|
24
|
Das S, Angsantikul P, Le C, Bao D, Miyamoto Y, Gao W, Zhang L, Eckmann L. Neutralization of cholera toxin with nanoparticle decoys for treatment of cholera. PLoS Negl Trop Dis 2018; 12:e0006266. [PMID: 29470490 PMCID: PMC5839590 DOI: 10.1371/journal.pntd.0006266] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 03/06/2018] [Accepted: 01/24/2018] [Indexed: 01/16/2023] Open
Abstract
Diarrheal diseases are a major cause of morbidity and mortality worldwide. In many cases, antibiotic therapy is either ineffective or not recommended due to concerns about emergence of resistance. The pathogenesis of several of the most prevalent infections, including cholera and enteroxigenic Escherichia coli, is dominated by enterotoxins produced by lumen-dwelling pathogens before clearance by intestinal defenses. Toxins gain access to the host through critical host receptors, making these receptors attractive targets for alternative antimicrobial strategies that do not rely on conventional antibiotics. Here, we developed a new nanotechnology strategy as a countermeasure against cholera, one of the most important and prevalent toxin-mediated enteric infections. The key host receptor for cholera toxin, monosialotetrahexosylganglioside (GM1), was coated onto the surface of polymeric nanoparticles. The resulting GM1-polymer hybrid nanoparticles were shown to function as toxin decoys by selectively and stably binding cholera toxin, and neutralizing its actions on epithelial cells in vitro and in vivo. Furthermore, the GM1-coated nanoparticle decoys attenuated epithelial 3’,5’-cyclic adenosine monophosphate production and fluid responses to infection with live Vibrio cholera in cell culture and a murine infection model. Together, these studies illustrate that the new nanotechnology-based platform can be employed as a non-traditional antimicrobial strategy for the management of enteric infections with enterotoxin-producing pathogens. Diarrheal diseases are a major cause of suffering and death in the world, particularly in tropical regions with limited health care resources. Many of the most important diarrhea-causing microbes produce toxins that activate fluid secretion in the gut. A prototype pathogen in this category is the cause of cholera, Vibrio cholerae, which is characterized by profuse diarrhea and severe electrolyte disturbances due to the release of cholera toxin. Although treatment with fluids by mouth or injection can save patients from death, they still experience the devastating symptoms of the disease. In the present study, we have developed a new intervention strategy with engineered nanoparticles, particulates than are smaller than one millionth of a meter, which can neutralize cholera toxin in the gut before it can cause the characteristic disease manifestations. This strategy represents a novel interventional approach whose mechanism of action is different from currently existing therapies, thus significantly broadening the medical armamentarium against cholera and perhaps other gut infections that cause diseases dominated by toxin production.
Collapse
Affiliation(s)
- Soumita Das
- Department of Pathology, University of California, San Diego, La Jolla, California, United States of America
| | - Pavimol Angsantikul
- Department of Nanoengineering, University of California, San Diego, La Jolla, California, United States of America
| | - Christine Le
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Denny Bao
- Department of Pathology, University of California, San Diego, La Jolla, California, United States of America
| | - Yukiko Miyamoto
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Weiwei Gao
- Department of Nanoengineering, University of California, San Diego, La Jolla, California, United States of America
| | - Liangfang Zhang
- Department of Nanoengineering, University of California, San Diego, La Jolla, California, United States of America
- * E-mail: (LE); (LZ)
| | - Lars Eckmann
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
- * E-mail: (LE); (LZ)
| |
Collapse
|
25
|
Ball RL, Bajaj P, Whitehead KA. Oral delivery of siRNA lipid nanoparticles: Fate in the GI tract. Sci Rep 2018; 8:2178. [PMID: 29391566 PMCID: PMC5794865 DOI: 10.1038/s41598-018-20632-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/22/2018] [Indexed: 01/28/2023] Open
Abstract
Oral delivery, a patient-friendly means of drug delivery, is preferred for local administration of intestinal therapeutics. Lipidoid nanoparticles, which have been previously shown to deliver siRNA to intestinal epithelial cells, have potential to treat intestinal disease. It is unknown, however, whether the oral delivery of these particles is possible. To better understand the fate of lipid nanoparticles in the gastrointestinal (GI) tract, we studied delivery under deconstructed stomach and intestinal conditions in vitro. Lipid nanoparticles remained potent and stable following exposure to solutions with pH values as low as 1.2. Efficacy decreased following exposure to “fed”, but not “fasting” concentrations of pepsin and bile salts. The presence of mucin on Caco-2 cells also reduced potency, although this effect was mitigated slightly by increasing the percentage of PEG in the lipid nanoparticle. Mouse biodistribution studies indicated that siRNA-loaded nanoparticles were retained in the GI tract for at least 8 hours. Although gene silencing was not initially observed following oral LNP delivery, confocal microscopy confirmed that nanoparticles entered the epithelial cells of the mouse small intestine and colon. Together, these data suggest that orally-delivered LNPs should be protected in the stomach and upper intestine to promote siRNA delivery to intestinal epithelial cells.
Collapse
Affiliation(s)
- Rebecca L Ball
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Palak Bajaj
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States.,Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States. .,Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States.
| |
Collapse
|
26
|
Lee Y, Thompson DH. Stimuli-responsive liposomes for drug delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:10.1002/wnan.1450. [PMID: 28198148 PMCID: PMC5557698 DOI: 10.1002/wnan.1450] [Citation(s) in RCA: 240] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/23/2016] [Accepted: 11/27/2016] [Indexed: 12/25/2022]
Abstract
The ultimate goal of drug delivery is to increase the bioavailability and reduce the toxic side effects of the active pharmaceutical ingredient (API) by releasing them at a specific site of action. In the case of antitumor therapy, association of the therapeutic agent with a carrier system can minimize damage to healthy, nontarget tissues, while limit systemic release and promoting long circulation to enhance uptake at the cancerous site due to the enhanced permeation and retention effect (EPR). Stimuli-responsive systems have become a promising way to deliver and release payloads in a site-selective manner. Potential carrier systems have been derived from a wide variety of materials, including inorganic nanoparticles, lipids, and polymers that have been imbued with stimuli-sensitive properties to accomplish triggered release based on an environmental cue. The unique features in the tumor microenvironment can serve as an endogenous stimulus (pH, redox potential, or unique enzymatic activity) or the locus of an applied external stimulus (heat or light) to trigger the controlled release of API. In liposomal carrier systems triggered release is generally based on the principle of membrane destabilization from local defects within bilayer membranes to effect release of liposome-entrapped drugs. This review focuses on the literature appearing between November 2008-February 2016 that reports new developments in stimuli-sensitive liposomal drug delivery strategies using pH change, enzyme transformation, redox reactions, and photochemical mechanisms of activation. WIREs Nanomed Nanobiotechnol 2017, 9:e1450. doi: 10.1002/wnan.1450 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Y Lee
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - D H Thompson
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
27
|
Giri TK, Bhowmick S, Maity S. Entrapment of capsaicin loaded nanoliposome in pH responsive hydrogel beads for colonic delivery. J Drug Deliv Sci Technol 2017. [DOI: 10.1016/j.jddst.2017.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
28
|
Ji Y, Lemberg M, Prudic A, Paus R, Sadowski G. Modeling and analysis of dissolution of paracetamol/Eudragit ® formulations. Chem Eng Res Des 2017. [DOI: 10.1016/j.cherd.2017.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Alavi S, Haeri A, Dadashzadeh S. Utilization of chitosan-caged liposomes to push the boundaries of therapeutic delivery. Carbohydr Polym 2017; 157:991-1012. [DOI: 10.1016/j.carbpol.2016.10.063] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 11/25/2022]
|
30
|
Nguyen TX, Huang L, Gauthier M, Yang G, Wang Q. Recent advances in liposome surface modification for oral drug delivery. Nanomedicine (Lond) 2016; 11:1169-85. [DOI: 10.2217/nnm.16.9] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Oral delivery via the gastrointestinal (GI) tract is the dominant route for drug administration. Orally delivered liposomal carriers can enhance drug solubility and protect the encapsulated theraputic agents from the extreme conditions found in the GI tract. Liposomes, with their fluid lipid bilayer membrane and their nanoscale size, can significantly improve oral absorption. Unfortunately, the clinical applications of conventional liposomes have been hindered due to their poor stability and availability under the harsh conditions typically presented in the GI tract. To overcome this problem, the surface modification of liposomes has been investigated. Although liposome surface modification has been extensively studied for oral drug delivery, no review exists so far that adequately covers this topic. The purpose of this paper is to summarize and critically analyze emerging trends in liposome surface modification for oral drug delivery.
Collapse
Affiliation(s)
- Thanh Xuan Nguyen
- Department of Biomedical Engineering, College of Life Science & Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- National Engineering Research Center for Nano-Medicine, Huazhong University of Science & Technology, Wuhan 430074, China
- Department of Human & Animal Physiology, Faculty of Biology-Agricultural Technology, Hanoi Pedagogical University No.2, Vietnam
| | - Lin Huang
- Department of Biomedical Engineering, College of Life Science & Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- National Engineering Research Center for Nano-Medicine, Huazhong University of Science & Technology, Wuhan 430074, China
- Wuhan East Lake High-tech Zone Administrative Committee, Wuhan 430079, China
| | - Mario Gauthier
- Department of Chemistry, University of Waterloo, 200 University Ave West, Waterloo, N2L 3G1, Canada
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science & Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- National Engineering Research Center for Nano-Medicine, Huazhong University of Science & Technology, Wuhan 430074, China
| | - Qun Wang
- Department of Chemical & Biological Engineering, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
31
|
Chitosan coatings to control release and target tissues for therapeutic delivery. Ther Deliv 2015; 6:855-71. [DOI: 10.4155/tde.15.31] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The natural biopolymer chitosan has versatile applications in therapeutic delivery. Coating drug delivery matrices or biomaterials with chitosan offers several advantages in drug delivery, including control of drug release, slowing degradation rate and improving biocompatibility. Advanced uses of chitosan in coating form include targeting drug delivery vehicles to specific tissue as well as providing a stimulus-controlled release response. The present review summarizes the current applications of chitosan coatings in the context of different biomaterial delivery technologies, as well as future directions of chitosan coatings for drug delivery technologies under development.
Collapse
|
32
|
Hua S, Marks E, Schneider JJ, Keely S. Advances in oral nano-delivery systems for colon targeted drug delivery in inflammatory bowel disease: selective targeting to diseased versus healthy tissue. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1117-32. [PMID: 25784453 DOI: 10.1016/j.nano.2015.02.018] [Citation(s) in RCA: 327] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 02/02/2015] [Accepted: 02/25/2015] [Indexed: 12/15/2022]
Abstract
UNLABELLED Colon targeted drug delivery is an active area of research for local diseases affecting the colon, as it improves the efficacy of therapeutics and enables localized treatment, which reduces systemic toxicity. Targeted delivery of therapeutics to the colon is particularly advantageous for the treatment of inflammatory bowel disease (IBD), which includes ulcerative colitis and Crohn's disease. Advances in oral drug delivery design have significantly improved the bioavailability of drugs to the colon; however in order for a drug to have therapeutic efficacy during disease, considerations must be made for the altered physiology of the gastrointestinal (GI) tract that is associated with GI inflammation. Nanotechnology has been used in oral dosage formulation design as strategies to further enhance uptake into diseased tissue within the colon. This review will describe some of the physiological challenges faced by orally administered delivery systems in IBD, the important developments in orally administered nano-delivery systems for colon targeting, and the future advances of this research. FROM THE CLINICAL EDITOR Inflammatory Bowel Disease (IBD) poses a significant problem for a large number of patients worldwide. Current medical therapy mostly aims at suppressing the active inflammatory episodes. In this review article, the authors described and discussed the various approaches current nano-delivery systems can offer in overcoming the limitations of conventional drug formulations.
Collapse
Affiliation(s)
- Susan Hua
- The School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.
| | - Ellen Marks
- The School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia; Gastrointestinal Research Group, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jennifer J Schneider
- The School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Simon Keely
- The School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia; Gastrointestinal Research Group, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
33
|
Hua S. Orally administered liposomal formulations for colon targeted drug delivery. Front Pharmacol 2014; 5:138. [PMID: 24959147 PMCID: PMC4050429 DOI: 10.3389/fphar.2014.00138] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/22/2014] [Indexed: 01/05/2023] Open
Affiliation(s)
- Susan Hua
- School of Biomedical Sciences and Pharmacy, The University of Newcastle Callaghan, NSW, Australia
| |
Collapse
|
34
|
Preparation of Starch/Gelatin Blend Microparticles by a Water-in-Oil Emulsion Method for Controlled Release Drug Delivery. Int J Biomater 2014; 2014:829490. [PMID: 24868207 PMCID: PMC4020388 DOI: 10.1155/2014/829490] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/06/2014] [Accepted: 04/08/2014] [Indexed: 11/29/2022] Open
Abstract
Information on the preparation and properties of starch/gelatin blend microparticles with and without crosslinking for drug delivery is presented. The blend microparticles were prepared by the water-in-oil emulsion solvent diffusion method. Glutaraldehyde and methylene blue were used as the crosslinker and the water-soluble drug model, respectively. The blend microparticles were characterized by scanning electron microscopy (SEM), Fourier transform infrared (FTIR) spectroscopy, and UV-Vis spectroscopy. The functional groups of the starch and gelatin blend matrices were determined from the FTIR spectra. Blend microparticles with a nearly spherical shape and internal porous structure were observed from SEM images. The average particle size of the gelatin microparticles depended on the crosslinker ratio but not on the starch/gelatin blend ratio. The in vitro drug release content significantly decreased as the crosslinker ratio increased and the starch blend ratio decreased. The results demonstrated that the starch/gelatin blend microparticles should be a useful controlled release delivery carrier for water-soluble drugs.
Collapse
|