1201
|
Ristori S, Grillo I, Lusa S, Thamm J, Valentino G, Campani V, Caraglia M, Steiniger F, Luciani P, De Rosa G. Structural Characterization of Self-Assembling Hybrid Nanoparticles for Bisphosphonate Delivery in Tumors. Mol Pharm 2018; 15:1258-1265. [DOI: 10.1021/acs.molpharmaceut.7b01085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Sandra Ristori
- Department of Chemistry, Center for Colloid and Surface Science (CSGI), via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Isabelle Grillo
- Large Scale Structures Group, Institut Laue Langevin, 71 rue des Martyrs, CS 20156, 38042 Grenoble Cedex 9, France
| | - Sara Lusa
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania “Luigi Vanvitelli”, Via S.M. Costantinopoli, 16, 80138 Naples, Italy
| | - Jana Thamm
- Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University, Lessingstrasse 8, 07743 Jena, Germany
| | - Gina Valentino
- Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University, Lessingstrasse 8, 07743 Jena, Germany
| | - Virginia Campani
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania “Luigi Vanvitelli”, Via S.M. Costantinopoli, 16, 80138 Naples, Italy
| | - Frank Steiniger
- Electron Microscopy Center, University Hospital Jena, Friedrich Schiller University, Ziegelmühlenweg 1, 07743 Jena, Germany
| | - Paola Luciani
- Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University, Lessingstrasse 8, 07743 Jena, Germany
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| |
Collapse
|
1202
|
Le PN, Huynh CK, Tran NQ. Advances in thermosensitive polymer-grafted platforms for biomedical applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:1016-1030. [PMID: 30184725 DOI: 10.1016/j.msec.2018.02.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 12/16/2017] [Accepted: 02/08/2018] [Indexed: 02/06/2023]
Abstract
Studies on "smart" polymeric material performing environmental stimuli such as temperature, pH, magnetic field, enzyme and photo-sensation have recently paid much attention to practical applications. Among of them, thermo-responsive grafted copolymers, amphiphilic steroids as well as polyester molecules have been utilized in the fabrication of several multifunctional platforms. Indeed, they performed a strikingly functional improvement comparing to some original materials and exhibited a holistic approach for biomedical applications. In case of drug delivery systems (DDS), there has been some successful proof of thermal-responsive grafted platforms on clinical trials such as ThermoDox®, BIND-014, Cynviloq IG-001, Genexol-PM, etc. This review would detail the recent progress and highlights of some temperature-responsive polymer-grafted nanomaterials or hydrogels in the 'smart' DDS that covered from synthetic polymers to nature-driven biomaterials and novel generations of some amphiphilic functional platforms. These approaches could produce several types of smart biomaterials for human health care in future.
Collapse
Affiliation(s)
- Phung Ngan Le
- Institute of Research and Development, Duy Tan University, Da Nang City 550000, Viet Nam; Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 1A TL29, District 12, Hochiminh City 700000, Viet Nam
| | - Chan Khon Huynh
- Biomedical Engineering Department, International University, National Universities in HCMC, HCMC 70000, Viet Nam
| | - Ngoc Quyen Tran
- Institute of Research and Development, Duy Tan University, Da Nang City 550000, Viet Nam; Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 1A TL29, District 12, Hochiminh City 700000, Viet Nam; Graduate School of Science and Technology Viet Nam, Vietnam Academy of Science and Technology, 1A TL29, District 12, Hochiminh City 700000, Viet Nam.
| |
Collapse
|
1203
|
Pashirova TN, Burilova EA, Lukashenko SS, Lenina OA, Zobov VV, Khamatgalimov AR, Kovalenko VI, Zakharova LY, Sinyashin OG. Synthesis, Self-Association, and Solubilizing Ability of an Amphiphilic Derivative of Poly(ethylene glycol) Methyl Ether. RUSS J GEN CHEM+ 2018. [DOI: 10.1134/s107036321712012x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
1204
|
Nisini R, Poerio N, Mariotti S, De Santis F, Fraziano M. The Multirole of Liposomes in Therapy and Prevention of Infectious Diseases. Front Immunol 2018; 9:155. [PMID: 29459867 PMCID: PMC5807682 DOI: 10.3389/fimmu.2018.00155] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/17/2018] [Indexed: 12/17/2022] Open
Abstract
Liposomes are closed bilayer structures spontaneously formed by hydrated phospholipids that are widely used as efficient delivery systems for drugs or antigens, due to their capability to encapsulate bioactive hydrophilic, amphipathic, and lipophilic molecules into inner water phase or within lipid leaflets. The efficacy of liposomes as drug or antigen carriers has been improved in the last years to ameliorate pharmacokinetics and capacity to release their cargo in selected target organs or cells. Moreover, different formulations and variations in liposome composition have been often proposed to include immunostimulatory molecules, ligands for specific receptors, or stimuli responsive compounds. Intriguingly, independent research has unveiled the capacity of several phospholipids to play critical roles as intracellular messengers in modulating both innate and adaptive immune responses through various mechanisms, including (i) activation of different antimicrobial enzymatic pathways, (ii) driving the fusion–fission events between endosomes with direct consequences to phagosome maturation and/or to antigen presentation pathway, and (iii) modulation of the inflammatory response. These features can be exploited by including selected bioactive phospholipids in the bilayer scaffold of liposomes. This would represent an important step forward since drug or antigen carrying liposomes could be engineered to simultaneously activate different signal transduction pathways and target specific cells or tissues to induce antigen-specific T and/or B cell response. This lipid-based host-directed strategy can provide a focused antimicrobial innate and adaptive immune response against specific pathogens and offer a novel prophylactic or therapeutic option against chronic, recurrent, or drug-resistant infections.
Collapse
Affiliation(s)
- Roberto Nisini
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Noemi Poerio
- Dipartimento di Biologia, Università degli Studi di Roma "Tor Vergata", Rome, Italy
| | - Sabrina Mariotti
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Federica De Santis
- Dipartimento di Biologia, Università degli Studi di Roma "Tor Vergata", Rome, Italy
| | - Maurizio Fraziano
- Dipartimento di Biologia, Università degli Studi di Roma "Tor Vergata", Rome, Italy
| |
Collapse
|
1205
|
Montenegro L, Turnaturi R, Parenti C, Pasquinucci L. Idebenone: Novel Strategies to Improve Its Systemic and Local Efficacy. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E87. [PMID: 29401722 PMCID: PMC5853719 DOI: 10.3390/nano8020087] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/27/2018] [Accepted: 01/30/2018] [Indexed: 12/16/2022]
Abstract
The key role of antioxidants in treating and preventing many systemic and topical diseases is well recognized. One of the most potent antioxidants available for pharmaceutical and cosmetic use is Idebenone (IDE), a synthetic analogue of Coenzyme Q10. Unfortunately, IDE's unfavorable physicochemical properties such as poor water solubility and high lipophilicity impair its bioavailability after oral and topical administration and prevent its parenteral use. In recent decades, many strategies have been proposed to improve IDE effectiveness in the treatment of neurodegenerative diseases and skin disorders. After a brief description of IDE potential therapeutic applications and its pharmacokinetic and pharmacodynamic profile, this review will focus on the different approaches investigated to overcome IDE drawbacks, such as IDE incorporation into different types of delivery systems (liposomes, cyclodextrins, microemulsions, self-micro-emulsifying drug delivery systems, lipid-based nanoparticles, polymeric nanoparticles) and IDE chemical modification. The results of these studies will be illustrated with emphasis on the most innovative strategies and their future perspectives.
Collapse
Affiliation(s)
- Lucia Montenegro
- Department of Drug Sciences, Pharmaceutical Technology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| | - Rita Turnaturi
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| | - Carmela Parenti
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| | - Lorella Pasquinucci
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| |
Collapse
|
1206
|
Dou XQ, Wang H, Zhang J, Wang F, Xu GL, Xu CC, Xu HH, Xiang SS, Fu J, Song HF. Aptamer-drug conjugate: targeted delivery of doxorubicin in a HER3 aptamer-functionalized liposomal delivery system reduces cardiotoxicity. Int J Nanomedicine 2018; 13:763-776. [PMID: 29440899 PMCID: PMC5804143 DOI: 10.2147/ijn.s149887] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Introduction The toxic side effects of doxorubicin (DOX) have limited its use in chemotherapy. Neither liposomal DOX nor pegylated liposomal DOX are able to completely resolve this issue. This is a proof-of-concept study testing aptamer-drug conjugate (ApDC) targeted delivery systems for chemotherapeutic drugs. Methods Aptamer library targeting human epidermal growth factor receptor 3 (HER3) was screened and affinity was determined by enzyme-linked immunosorbent assay. Specificity was tested in MCF-7HER3-high, BT474HER3-high, and 293THER3-negative cells using flow cytometry and confocal microscopy. We further developed a HER3 aptamer-functionalized liposome encapsulating DOX and the efficiency of this ApDC was detected by cellular uptake analysis and cell viability assay. In MCF-7 tumor-bearing mice, tumor targeting evaluation, efficacy, toxicity and preliminary pharmocokinetic study was performed. Results The candidate #13 aptamer had highest affinity (Kd =98±9.7 nM) and specificity. ApDC effectively reduces the half maximal inhibitory concentration of DOX compared with lipsome-DOX and free DOX. In vivo imaging and preliminary distribution studies showed that actively targeted nanoparticles, such as Apt-Lip-DOX molecules, could facilitate the delivery of DOX into tumors in MCF-7-bearing mice. This targeted chemotherapy caused greater tumor suppression than other groups and alleviated side effects such as weight loss, low survival rate, and organ (heart and liver) injury demonstrated by H&E staining. Conclusion The results indicate that targeted chemotherapy using the aptamer-drug conjugate format could provide better tolerability and efficacy compared with non-targeted delivery in relatively low-dose toxic drugs.
Collapse
Affiliation(s)
- Xiao-Qian Dou
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hua Wang
- Ophthalmology Department, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Zhang
- Bioanalysis Department, United-Power Pharma Tech Co., Ltd., Beijing, China
| | - Fang Wang
- Bioanalysis Department, United-Power Pharma Tech Co., Ltd., Beijing, China
| | - Gui-Li Xu
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Cheng-Cheng Xu
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Huan-Hua Xu
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Shen-Si Xiang
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jie Fu
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hai-Feng Song
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
1207
|
Gonçalves A, Nikmaram N, Roohinejad S, Estevinho BN, Rocha F, Greiner R, McClements DJ. Production, properties, and applications of solid self-emulsifying delivery systems (S-SEDS) in the food and pharmaceutical industries. Colloids Surf A Physicochem Eng Asp 2018. [DOI: 10.1016/j.colsurfa.2017.10.076] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
1208
|
Kurabi A, Schaerer D, Chang L, Pak K, Ryan AF. Optimisation of peptides that actively cross the tympanic membrane by random amino acid extension: a phage display study. J Drug Target 2018; 26:127-134. [PMID: 28658990 PMCID: PMC6223256 DOI: 10.1080/1061186x.2017.1347791] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 06/24/2017] [Indexed: 10/19/2022]
Abstract
Local treatment of middle ear (ME) disease currently requires surgical penetration of the tympanic membrane (TM). We previously discovered 12-mer peptides that are actively transported across the intact TM, a process that could be used for non-invasive drug delivery into the ME. To optimise transport and provide further understanding of the peptides transport mechanism, we extended two of the candidate peptides by six additional amino acids at random, and screened the resulting 18-mers libraries on TMs of rats with active bacterial otitis media (OM) for transport efficiency using phage display. Six identified peptides were individually tested in vivo for trans-TM transport to verify the tissue specificity. Three exhibited enhanced transport compared to their parent 12-mer scaffold, with the best showing an approximately nine-fold increase. Sequence analysis revealed anchor residues and structural features associated with enhanced transport. This included the prominent display of conserved sequence motifs at the extended free ends of the predicted peptide structures.
Collapse
Affiliation(s)
- Arwa Kurabi
- a Department of Surgery, Division of Otolaryngology , School of Medicine, University of California , La Jolla , CA , USA
- b San Diego VA Healthcare System , San Diego , CA , USA
| | - Daniel Schaerer
- a Department of Surgery, Division of Otolaryngology , School of Medicine, University of California , La Jolla , CA , USA
| | - Lisa Chang
- a Department of Surgery, Division of Otolaryngology , School of Medicine, University of California , La Jolla , CA , USA
| | - Kwang Pak
- a Department of Surgery, Division of Otolaryngology , School of Medicine, University of California , La Jolla , CA , USA
- b San Diego VA Healthcare System , San Diego , CA , USA
| | - Allen F Ryan
- a Department of Surgery, Division of Otolaryngology , School of Medicine, University of California , La Jolla , CA , USA
- b San Diego VA Healthcare System , San Diego , CA , USA
| |
Collapse
|
1209
|
Ahmadzada T, Reid G, McKenzie DR. Fundamentals of siRNA and miRNA therapeutics and a review of targeted nanoparticle delivery systems in breast cancer. Biophys Rev 2018; 10:69-86. [PMID: 29327101 PMCID: PMC5803180 DOI: 10.1007/s12551-017-0392-1] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 12/15/2017] [Indexed: 12/11/2022] Open
Abstract
Gene silencing via RNA interference (RNAi) is rapidly evolving as a personalized approach to cancer treatment. The effector molecules-small interfering RNAs (siRNAs) and microRNAs (miRNAs)-can be used to silence or "switch off" specific cancer genes. Currently, the main barrier to implementing siRNA- and miRNA-based therapies in clinical practice is the lack of an effective delivery system that can protect the RNA molecules from nuclease degradation, deliver to them to tumor tissue, and release them into the cytoplasm of the target cancer cells, all without inducing adverse effects. Here, we review the fundamentals of RNAi, cell membrane transport pathways, and factors that affect intracellular delivery. We discuss the advantages and disadvantages of the various types of nanoparticle delivery systems, with a focus on those that have been investigated in breast cancer in vivo.
Collapse
Affiliation(s)
- Tamkin Ahmadzada
- Sydney Medical School, The University of Sydney, Sydney, Australia.
| | - Glen Reid
- Sydney Medical School, The University of Sydney, Sydney, Australia
- Asbestos Diseases Research Institute (ADRI), Sydney, Australia
| | | |
Collapse
|
1210
|
Lamichhane N, Udayakumar TS, D'Souza WD, Simone CB, Raghavan SR, Polf J, Mahmood J. Liposomes: Clinical Applications and Potential for Image-Guided Drug Delivery. Molecules 2018; 23:molecules23020288. [PMID: 29385755 PMCID: PMC6017282 DOI: 10.3390/molecules23020288] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 01/16/2023] Open
Abstract
Liposomes have been extensively studied and are used in the treatment of several diseases. Liposomes improve the therapeutic efficacy by enhancing drug absorption while avoiding or minimizing rapid degradation and side effects, prolonging the biological half-life and reducing toxicity. The unique feature of liposomes is that they are biocompatible and biodegradable lipids, and are inert and non-immunogenic. Liposomes can compartmentalize and solubilize both hydrophilic and hydrophobic materials. All these properties of liposomes and their flexibility for surface modification to add targeting moieties make liposomes more attractive candidates for use as drug delivery vehicles. There are many novel liposomal formulations that are in various stages of development, to enhance therapeutic effectiveness of new and established drugs that are in preclinical and clinical trials. Recent developments in multimodality imaging to better diagnose disease and monitor treatments embarked on using liposomes as diagnostic tool. Conjugating liposomes with different labeling probes enables precise localization of these liposomal formulations using various modalities such as PET, SPECT, and MRI. In this review, we will briefly review the clinical applications of liposomal formulation and their potential imaging properties.
Collapse
Affiliation(s)
- Narottam Lamichhane
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | - Warren D D'Souza
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Charles B Simone
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Srinivasa R Raghavan
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA.
| | - Jerimy Polf
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Javed Mahmood
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
1211
|
Delgado JL, Hsieh CM, Chan NL, Hiasa H. Topoisomerases as anticancer targets. Biochem J 2018; 475:373-398. [PMID: 29363591 PMCID: PMC6110615 DOI: 10.1042/bcj20160583] [Citation(s) in RCA: 295] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/14/2017] [Accepted: 12/21/2017] [Indexed: 12/15/2022]
Abstract
Many cancer type-specific anticancer agents have been developed and significant advances have been made toward precision medicine in cancer treatment. However, traditional or nonspecific anticancer drugs are still important for the treatment of many cancer patients whose cancers either do not respond to or have developed resistance to cancer-specific anticancer agents. DNA topoisomerases, especially type IIA topoisomerases, are proved therapeutic targets of anticancer and antibacterial drugs. Clinically successful topoisomerase-targeting anticancer drugs act through topoisomerase poisoning, which leads to replication fork arrest and double-strand break formation. Unfortunately, this unique mode of action is associated with the development of secondary cancers and cardiotoxicity. Structures of topoisomerase-drug-DNA ternary complexes have revealed the exact binding sites and mechanisms of topoisomerase poisons. Recent advances in the field have suggested a possibility of designing isoform-specific human topoisomerase II poisons, which may be developed as safer anticancer drugs. It may also be possible to design catalytic inhibitors of topoisomerases by targeting certain inactive conformations of these enzymes. Furthermore, identification of various new bacterial topoisomerase inhibitors and regulatory proteins may inspire the discovery of novel human topoisomerase inhibitors. Thus, topoisomerases remain as important therapeutic targets of anticancer agents.
Collapse
Affiliation(s)
- Justine L Delgado
- Division of Medicinal and Natural Products Chemistry, Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 115 S Grand Ave., S321 Pharmacy Building, Iowa City, IA 52242, U.S.A
| | - Chao-Ming Hsieh
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei City 100, Taiwan
| | - Nei-Li Chan
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei City 100, Taiwan
| | - Hiroshi Hiasa
- Department of Pharmacology, University of Minnesota Medical School, 6-120 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, U.S.A.
| |
Collapse
|
1212
|
Cholesterol modulates the liposome membrane fluidity and permeability for a hydrophilic molecule. Food Chem Toxicol 2018; 113:40-48. [PMID: 29337230 DOI: 10.1016/j.fct.2018.01.017] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 01/27/2023]
Abstract
The effect of cholesterol (CHOL) content on the permeability and fluidity of dipalmitoylphosphatidylcholine (DPPC) liposome membrane was investigated. Liposomes encapsulating sulforhodamine B (SRB), a fluorescent dye, were prepared by reverse phase evaporation technique (REV) at various DPPC:CHOL molar ratios (from 100:0 to 100:100). The release kinetics of SRB was studied during 48 h in buffer (pH 7.4) containing NaCl at 37 °C. The DPPC:CHOL formulations were also characterized for their size, polydispersity index and morphology. Increasing CHOL concentration induced an increase in the mean liposomes size accompanying with a shape transition from irregular to nanosized, regular and spherical vesicles. The release kinetics of SRB showed a biphasic pattern; the release data was then analyzed using different mathematical models. On the overall, the SRB release was governed by a non-Fickian diffusion during the first period (0-10 h) while it followed a Fickian diffusion between 10 and 48 h. Changes in DPPC liposome membrane fluidity of various batches (CHOL% 0, 10, 20, 30 and 100) were monitored by using 5- and 16 doxyl stearic acids (DSA) as spin labels. CHOL induced a decrease in the bilayer fluidity. Concisely, CHOL represents a critical component in modulating the release of hydrophilic molecules from lipid vesicles.
Collapse
|
1213
|
Riaz MK, Riaz MA, Zhang X, Lin C, Wong KH, Chen X, Zhang G, Lu A, Yang Z. Surface Functionalization and Targeting Strategies of Liposomes in Solid Tumor Therapy: A Review. Int J Mol Sci 2018; 19:E195. [PMID: 29315231 PMCID: PMC5796144 DOI: 10.3390/ijms19010195] [Citation(s) in RCA: 284] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/03/2018] [Accepted: 01/04/2018] [Indexed: 12/23/2022] Open
Abstract
Surface functionalization of liposomes can play a key role in overcoming the current limitations of nanocarriers to treat solid tumors, i.e., biological barriers and physiological factors. The phospholipid vesicles (liposomes) containing anticancer agents produce fewer side effects than non-liposomal anticancer formulations, and can effectively target the solid tumors. This article reviews information about the strategies for targeting of liposomes to solid tumors along with the possible targets in cancer cells, i.e., extracellular and intracellular targets and targets in tumor microenvironment or vasculature. Targeting ligands for functionalization of liposomes with relevant surface engineering techniques have been described. Stimuli strategies for enhanced delivery of anticancer agents at requisite location using stimuli-responsive functionalized liposomes have been discussed. Recent approaches for enhanced delivery of anticancer agents at tumor site with relevant surface functionalization techniques have been reviewed. Finally, current challenges of functionalized liposomes and future perspective of smart functionalized liposomes have been discussed.
Collapse
Affiliation(s)
- Muhammad Kashif Riaz
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.
| | - Muhammad Adil Riaz
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Xue Zhang
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.
| | - Congcong Lin
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.
| | - Ka Hong Wong
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.
| | - Xiaoyu Chen
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.
| | - Ge Zhang
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.
| |
Collapse
|
1214
|
Mathiyazhakan M, Wiraja C, Xu C. A Concise Review of Gold Nanoparticles-Based Photo-Responsive Liposomes for Controlled Drug Delivery. NANO-MICRO LETTERS 2018; 10:10. [PMID: 30393659 PMCID: PMC6199057 DOI: 10.1007/s40820-017-0166-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/09/2017] [Indexed: 05/18/2023]
Abstract
The focus of drug delivery is shifting toward smart drug carriers that release the cargo in response to a change in the microenvironment due to an internal or external trigger. As the most clinically successful nanosystem, liposomes naturally come under the spotlight of this trend. This review summarizes the latest development about the design and construction of photo-responsive liposomes with gold nanoparticles for the controlled drug release. Alongside, we overview the mechanism involved in this process and the representative applications.
Collapse
Affiliation(s)
- Malathi Mathiyazhakan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
| | - Christian Wiraja
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
| | - Chenjie Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore.
| |
Collapse
|
1215
|
Campani V, Giarra S, De Rosa G. Lipid-based core-shell nanoparticles: Evolution and potentialities in drug delivery. OPENNANO 2018. [DOI: 10.1016/j.onano.2017.12.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
1216
|
|
1217
|
Nandwana V, Singh A, You MM, Zhang G, Higham J, Zheng TS, Li Y, Prasad PV, Dravid VP. Magnetic lipid nanocapsules (MLNCs): self-assembled lipid-based nanoconstruct for non-invasive theranostic applications. J Mater Chem B 2018; 6:1026-1034. [DOI: 10.1039/c7tb03160b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A novel magnetic nanostructures (MNS) stabilized lipid nanoconstruct is reported that shows superior structural stability and theranostic functionality than conventional lipid based nanocarriers.
Collapse
Affiliation(s)
- Vikas Nandwana
- Department of Materials Science & Engineering
- Northwestern University
- Evanston
- USA
- International Institute of Nanotechnology
| | - Abhalaxmi Singh
- Department of Materials Science & Engineering
- Northwestern University
- Evanston
- USA
- International Institute of Nanotechnology
| | - Marisa M. You
- Department of Materials Science & Engineering
- Northwestern University
- Evanston
- USA
| | - Gefei Zhang
- Department of Materials Science & Engineering
- Northwestern University
- Evanston
- USA
| | - John Higham
- Department of Materials Science & Engineering
- Northwestern University
- Evanston
- USA
- Department of Biomedical Engineering
| | - Tiffany S. Zheng
- Department of Materials Science & Engineering
- Northwestern University
- Evanston
- USA
| | - Yue Li
- Department of Materials Science & Engineering
- Northwestern University
- Evanston
- USA
| | | | - Vinayak P. Dravid
- Department of Materials Science & Engineering
- Northwestern University
- Evanston
- USA
- International Institute of Nanotechnology
| |
Collapse
|
1218
|
Rideau E, Dimova R, Schwille P, Wurm FR, Landfester K. Liposomes and polymersomes: a comparative review towards cell mimicking. Chem Soc Rev 2018; 47:8572-8610. [DOI: 10.1039/c8cs00162f] [Citation(s) in RCA: 521] [Impact Index Per Article: 74.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Minimal cells: we compare and contrast liposomes and polymersomes for a bettera priorichoice and design of vesicles and try to understand the advantages and shortcomings associated with using one or the other in many different aspects (properties, synthesis, self-assembly, applications).
Collapse
Affiliation(s)
- Emeline Rideau
- Max Planck Institute for Polymer Research
- 55128 Mainz
- Germany
| | - Rumiana Dimova
- Max Planck Institute for Colloids and Interfaces
- Wissenschaftspark Potsdam-Golm
- 14476 Potsdam
- Germany
| | - Petra Schwille
- Max Planck Institute of Biochemistry
- 82152 Martinsried
- Germany
| | | | | |
Collapse
|
1219
|
Aziz DE, Abdelbary AA, Elassasy AI. Implementing Central Composite Design for Developing Transdermal Diacerein-Loaded Niosomes: Ex vivo Permeation and In vivo Deposition. Curr Drug Deliv 2018; 15:1330-1342. [PMID: 29921206 PMCID: PMC6340157 DOI: 10.2174/1567201815666180619105419] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/07/2018] [Accepted: 06/08/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Niosomes are surfactant-based vesicular nanosystems that proved their efficiency in transdermal delivery by overcoming skin inherent anatomic barrier; startum corneum. Central composite design is an efficient tool for developing and optimizing niosomal formulations using fewer experiments. OBJECTIVE The objective of this study was to prepare niosomes as a transdermal delivery system of diacerein using film hydration technique, employing central composite design, for avoiding its oral gastrointestinal problems. METHODS Three-level three-factor central composite design was employed for attaining optimal niosomes formulation with the desired characteristics. Three formulation variables were assessed: amount of salt in hydration medium (X1), lipid amount (X2) and number of surfactant parts (X3). DCNloaded niosomes were evaluated for entrapment efficiency percent (Y1), particle size (Y2), polydispersity index (Y3) and zeta potential (Y4). The suggested optimal niosomes were subjected to further characterization and utilized as a nucleus for developing elastic vesicles for comparative ex vivo and in vivo studies. RESULTS The values of the independent variables (X1, X2 and X3) in the optimal niosomes formulation were 0 g, 150 mg and 5 parts, respectively. It showed entrapment efficiency percentage of 95.63%, particle size of 436.65 nm, polydispersity index of 0.47 and zeta potential of -38.80 mV. Results of ex vivo permeation and skin deposition studies showed enhanced skin permeation and retention capacity of the prepared vesicles than drug suspension. CONCLUSION Results revealed that a transdermal niosomal system was successfully prepared and evaluated using central composite design which will result in delivering diacerein efficiently, avoiding its oral problems.
Collapse
Affiliation(s)
| | - Aly Ahmed Abdelbary
- Address correspondence to this author at the Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini, 11562, Cairo, Egypt; Tel: +201149005526; E-mail:
| | | |
Collapse
|
1220
|
Huang JL, Chen HZ, Gao XL. Lipid-coated calcium phosphate nanoparticle and beyond: a versatile platform for drug delivery. J Drug Target 2017; 26:398-406. [PMID: 29258343 DOI: 10.1080/1061186x.2017.1419360] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In recent years, lipid-coated calcium-phosphate (LCP) nanoparticle has been developed as a versatile platform for delivery of various therapeutics including gene, protein/peptide, chemotherapeutics and theranostic agents. The high endosomal escape, coupled with the ability to efficiently encapsulate phosphorylated drugs or prodrugs, make LCP become attractive vehicle for drug delivery. Additionally, the principle behind LCP formulation has also allowed rational design of LCP-derived nanoparticles (NPs) with other solid core or lipid membrane to overcome the various drug delivery barriers. Here, we briefly review the history of the development of LCP NPs, highlight the optimisations and modulations in the development process, and summarise the major applications of LCP NPs and LCP-derived NPs in drug delivery.
Collapse
Affiliation(s)
- Jia-Lin Huang
- a Department of Pharmacology and Chemical Biology, Faculty of Basic Medicine , Shanghai Jiao Tong University School of Medicine , Shanghai , PR China.,b Department of Neurological Surgery , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , PR China
| | - Hong-Zhuan Chen
- a Department of Pharmacology and Chemical Biology, Faculty of Basic Medicine , Shanghai Jiao Tong University School of Medicine , Shanghai , PR China
| | - Xiao-Ling Gao
- a Department of Pharmacology and Chemical Biology, Faculty of Basic Medicine , Shanghai Jiao Tong University School of Medicine , Shanghai , PR China
| |
Collapse
|
1221
|
Andrés-Guerrero V, Bravo-Osuna I, Pastoriza P, Molina-Martinez IT, Herrero-Vanrell R. Novel technologies for the delivery of ocular therapeutics in glaucoma. J Drug Deliv Sci Technol 2017. [DOI: 10.1016/j.jddst.2017.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
1222
|
Rehman M, Ihsan A, Madni A, Bajwa SZ, Shi D, Webster TJ, Khan WS. Solid lipid nanoparticles for thermoresponsive targeting: evidence from spectrophotometry, electrochemical, and cytotoxicity studies. Int J Nanomedicine 2017; 12:8325-8336. [PMID: 29200845 PMCID: PMC5701611 DOI: 10.2147/ijn.s147506] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Thermoresponsive drug delivery systems are designed for the controlled and targeted release of therapeutic payload. These systems exploit hyperthermic temperatures (>39°C), which may be applied by some external means or due to an encountered symptom in inflammatory diseases such as cancer and arthritis. The objective of this paper was to provide some solid evidence in support of the hypothesis that solid lipid nanoparticles (SLNs) can be used for thermoresponsive targeting by undergoing solid–liquid phase transition at their melting point (MP). Thermoresponsive lipid mixtures were prepared by mixing solid and liquid natural fatty acids, and their MP was measured by differential scanning calorimetry (DSC). SLNs (MP 39°C) containing 5-fluorouracil (5-FU) were synthesized by hot melt encapsulation method, and were found to have spherical shape (transmission electron microscopy studies), desirable size (<200 nm), and enhanced physicochemical stability (Fourier transform infrared spectroscopy analysis). We observed a sustained release pattern (22%–34%) at 37°C (5 hours). On the other hand, >90% drug was released at 39°C after 5 hours, suggesting that the SLNs show thermoresponsive drug release, thus confirming our hypothesis. Drug release from SLNs at 39°C was similar to oleic acid and linoleic acid nanoemulsions used in this study, which further confirmed that thermoresponsive drug release is due to solid–liquid phase transition. Next, a differential pulse voltammetry-based electrochemical chemical detection method was developed for quick and real-time analysis of 5-FU release, which also confirmed thermoresponsive drug release behavior of SLNs. Blank SLNs were found to be biocompatible with human gingival fibroblast cells, although 5-FU-loaded SLNs showed some cytotoxicity after 24 hours. 5-FU-loaded SLNs showed thermoresponsive cytotoxicity to breast cancer cells (MDA-MB-231) as cytotoxicity was higher at 39°C (cell viability 72%–78%) compared to 37°C (cell viability >90%) within 1 hour. In conclusion, this study presents SLNs as a safe, simple, and effective platform for thermoresponsive targeting.
Collapse
Affiliation(s)
- Mubashar Rehman
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Punjab, Pakistan.,Nanobiotech Group, National Institute of Biotechnology and Genetic Engineering, Faisalabad, Punjab, Pakistan.,Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Ayesha Ihsan
- Nanobiotech Group, National Institute of Biotechnology and Genetic Engineering, Faisalabad, Punjab, Pakistan
| | - Asadullah Madni
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Punjab, Pakistan
| | - Sadia Zafar Bajwa
- Nanobiotech Group, National Institute of Biotechnology and Genetic Engineering, Faisalabad, Punjab, Pakistan
| | - Di Shi
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA.,Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Waheed S Khan
- Nanobiotech Group, National Institute of Biotechnology and Genetic Engineering, Faisalabad, Punjab, Pakistan
| |
Collapse
|
1223
|
De Serrano LO, Burkhart DJ. Liposomal vaccine formulations as prophylactic agents: design considerations for modern vaccines. J Nanobiotechnology 2017; 15:83. [PMID: 29149896 PMCID: PMC5693489 DOI: 10.1186/s12951-017-0319-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/09/2017] [Indexed: 01/04/2023] Open
Abstract
Vaccinology is one of the most important cornerstones in modern medicine, providing better quality of life. The human immune system is composed of innate and adaptive immune processes that interplay when infection occurs. Innate immunity relies on pathogen-associated molecular patterns which are recognized by pathogen recognition receptors localized in antigen presenting cells. After antigen processing and presentation, CD4+ T cell polarization occurs, further leading to B cell and CD8+ activation and humoral and cell-mediated adaptive immune responses. Liposomes are being employed as vaccine technologies and their design is of importance to ensure proper immune responses. Physicochemical parameters like liposome size, charge, lamellarity and bilayer fluidity must be completely understood to ensure optimal vaccine stability and efficacy. Liposomal vaccines can be developed to target specific immune cell types for the induction of certain immune responses. In this review, we will present promising liposomal vaccine approaches for the treatment of important viral, bacterial, fungal and parasitic infections (including tuberculosis, TB). Cationic liposomes are the most studied liposome types due to their enhanced interaction with the negatively charged immune cells. Thus, a special section on the cationic lipid dimethyldioctadecylammonium and TB is also presented.
Collapse
Affiliation(s)
- Luis O. De Serrano
- Department of Biomedical & Pharmaceutical Sciences and Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812 USA
| | - David J. Burkhart
- Department of Biomedical & Pharmaceutical Sciences and Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812 USA
| |
Collapse
|
1224
|
Rwei AY, Wang B, Ji T, Zhan C, Kohane DS. Enhanced Triggering of Local Anesthetic Particles by Photosensitization and Photothermal Effect Using a Common Wavelength. NANO LETTERS 2017; 17:7138-7145. [PMID: 29058443 PMCID: PMC7491648 DOI: 10.1021/acs.nanolett.7b04176] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
On-demand pain relief systems would be very helpful additions to the armamentarium of pain management. Near-infrared triggered drug delivery systems have demonstrated the potential to provide such care. However, challenges remain in making such systems as stimulus-sensitive as possible, to enhance depth of tissue penetration, repeatability of triggering, and safety. Here we developed liposomes containing the local anesthetic tetrodotoxin and also containing a photosensitizer and gold nanorods that were excitable at the same near-infrared wavelength. The combination of triggering mechanisms enhanced the photosensitivity and repeatability of the system in vitro when compared with liposomes with a single photoresponsive component. In vivo, on-demand local anesthesia could be induced with a low irradiance and short irradiation duration, and liposomes containing both photosensitizer and gold nanorods were more effective than those containing just one photoresponsive component. Tissue reaction was benign.
Collapse
Affiliation(s)
- Alina Y. Rwei
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce Wang
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tianjiao Ji
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Changyou Zhan
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel S. Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- To whom correspondence may be addressed. (D.S. Kohane)
| |
Collapse
|
1225
|
Shigehiro T, Masuda J, Saito S, Khayrani AC, Jinno K, Seno A, Vaidyanath A, Mizutani A, Kasai T, Murakami H, Satoh A, Ito T, Hamada H, Seno Y, Mandai T, Seno M. Practical Liposomal Formulation for Taxanes with Polyethoxylated Castor Oil and Ethanol with Complete Encapsulation Efficiency and High Loading Efficiency. NANOMATERIALS 2017; 7:nano7100290. [PMID: 28946623 PMCID: PMC5666455 DOI: 10.3390/nano7100290] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/13/2017] [Accepted: 09/19/2017] [Indexed: 12/17/2022]
Abstract
Taxanes including paclitaxel and docetaxel are effective anticancer agents preferably sufficient for liposomal drug delivery. However, the encapsulation of these drugs with effective amounts into conventional liposomes is difficult due to their high hydrophobicity. Therefore, an effective encapsulation strategy for liposomal taxanes has been eagerly anticipated. In this study, the mixture of polyethoxylated castor oil (Cremophor EL) and ethanol containing phosphate buffered saline termed as CEP was employed as a solvent of the inner hydrophilic core of liposomes where taxanes should be incorporated. Docetaxel-, paclitaxel-, or 7-oxacetylglycosylated paclitaxel-encapsulating liposomes were successfully prepared with almost 100% of encapsulation efficiency and 29.9, 15.4, or 29.1 mol% of loading efficiency, respectively. We then applied the docetaxel-encapsulating liposomes for targeted drug delivery. Docetaxel-encapsulating liposomes were successfully developed HER2-targeted drug delivery by coupling HER2-specific binding peptide on liposome surface. The HER2-targeting liposomes exhibited HER2-specific internalization and enhanced anticancer activity in vitro. Therefore, we propose the sophisticated preparation of liposomal taxanes using CEP as a promising formulation for effective cancer therapies.
Collapse
Affiliation(s)
- Tsukasa Shigehiro
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
- Japan Society for the Promotion of Science, Tokyo 102-0083, Japan.
| | - Junko Masuda
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| | - Shoki Saito
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| | - Apriliana C Khayrani
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| | - Kazumasa Jinno
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| | - Akimasa Seno
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| | - Arun Vaidyanath
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| | - Akifumi Mizutani
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| | - Tomonari Kasai
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| | - Hiroshi Murakami
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| | - Ayano Satoh
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| | - Tetsuya Ito
- Ensuiko Sugar Refining Co., Ltd., Tokyo 102-0083, Japan.
| | - Hiroki Hamada
- Faculty of Science, Okayama University of Science, Okayama 700-0082, Japan.
| | - Yuhki Seno
- Faculty of Life Science, Kurashiki University of Science and the Arts, Kurashiki 712-8505, Japan.
| | - Tadakatsu Mandai
- Faculty of Life Science, Kurashiki University of Science and the Arts, Kurashiki 712-8505, Japan.
| | - Masaharu Seno
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-0082, Japan.
| |
Collapse
|
1226
|
Guo X, Zhu X, Liu D, Gong Y, Sun J, Dong C. Continuous delivery of propranolol from liposomes-in-microspheres significantly inhibits infantile hemangioma growth. Int J Nanomedicine 2017; 12:6923-6936. [PMID: 29075111 PMCID: PMC5609781 DOI: 10.2147/ijn.s137634] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose To reduce the adverse effects and high frequency of administration of propranolol to treat infantile hemangioma, we first utilized propranolol-loaded liposomes-in-microsphere (PLIM) as a novel topical release system to realize sustained release of propranolol. Methods PLIM was developed from encapsulating propranolol-loaded liposomes (PLs) in microspheres made of poly(lactic-co-glycolic acid)-b-poly(ethylene glycol)-b-poly(lactic-co-glycolic acid) copolymers (PLGA-PEG-PLGA). The release profile of propranolol from PLIM was evaluated, and its biological activity was investigated in vitro using proliferation assays on hemangioma stem cells (HemSCs). Tumor inhibition was studied in nude mice bearing human subcutaneous infantile hemangioma. Results The microspheres were of desired particle size (~77.8 μm) and drug encapsulation efficiency (~23.9%) and achieved sustained drug release for 40 days. PLIM exerted efficient inhibition of the proliferation of HemSCs and significantly reduced the expression of two angiogenesis factors (vascular endothelial growth factor-A [VEGF-A] and basic fibroblast growth factor [bFGF]) in HemSCs. Notably, the therapeutic effect of PLIM in hemangioma was superior to that of propranolol and PL in vivo, as reflected by significantly reduced hemangioma volume, weight, and microvessel density. The mean hemangioma weight of the PLIM-treated group was significantly lower than that of other groups (saline =0.28 g, propranolol =0.21 g, PL =0.13 g, PLIM =0.03 g; PLIM vs saline: P<0.001, PLIM vs propranolol: P<0.001, PLIM vs PL: P<0.001). The mean microvessel density of the PLIM-treated group was significantly lower than that of other groups (saline =40 vessels/mm2, propranolol =31 vessels/mm2, PL =25 vessels/mm2, PLIM =11 vessels/mm2; PLIM vs saline: P<0.001, PLIM vs propranolol: P<0.01, PLIM vs PL: P<0.05). Conclusion Our findings show that PLIM is a very promising approach to locally and efficiently deliver propranolol to the hemangioma site leading to a significant inhibition of infantile hemangioma.
Collapse
Affiliation(s)
- Xiaonan Guo
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Xiaoshuang Zhu
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Dakan Liu
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Yubin Gong
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Jing Sun
- Department of Pharmacy, Second Military Medical University, Shanghai, People's Republic of China
| | - Changxian Dong
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| |
Collapse
|
1227
|
Sehnert B, Burkhardt H, Finzel S, Dübel S, Voll RE. The sneaking ligand approach for cell type-specific modulation of intracellular signalling pathways. Clin Immunol 2017; 186:14-20. [PMID: 28867254 DOI: 10.1016/j.clim.2017.08.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 11/30/2022]
Abstract
Small molecules interfering with intracellular signalling pathways are used in the treatment of multiple diseases including RA. However, small molecules usually affect signalling in most cell types, not only in those which need to be targeted. This general inhibition of signalling pathways causes often adverse effects, which could be avoided by cell type-specific inhibitors. For cell-type specific modulation of signal transduction, we developed the sneaking ligand fusion proteins (SLFPs). SLFPs contain three domains: (1) the binding domain mediating cell type-specific targeting and endocytosis; (2) the endosomal release sequence releasing the effector domain into the cytoplasm; (3) the effector domain modulating signalling. Using our SLFP NF-kappaB inhibitor termed SLC1 we demonstrated that cell-type-specific modulation of intracellular signalling pathways is feasible, that endothelial NF-kappaB activation is critical for arthritis and peritonitis and that SLFPs help to identify disease-relevant pathways in defined cell types. Hence, SLFPs may improve risk-benefit ratios of therapeutic interventions.
Collapse
Affiliation(s)
- Bettina Sehnert
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Harald Burkhardt
- Division of Rheumatology, Department of Internal Medicine II, Fraunhofer IME-Project-Group Translational Medicine and Pharmacology, Johann Wolfgang Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Stephanie Finzel
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Dübel
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technical University Braunschweig, Braunschweig, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
1228
|
Rufino-Ramos D, Albuquerque PR, Carmona V, Perfeito R, Nobre RJ, Pereira de Almeida L. Extracellular vesicles: Novel promising delivery systems for therapy of brain diseases. J Control Release 2017; 262:247-258. [DOI: 10.1016/j.jconrel.2017.07.001] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/30/2017] [Accepted: 07/01/2017] [Indexed: 12/11/2022]
|
1229
|
Novel Drug Delivery Systems Tailored for Improved Administration of Glucocorticoids. Int J Mol Sci 2017; 18:ijms18091836. [PMID: 28837059 PMCID: PMC5618485 DOI: 10.3390/ijms18091836] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/18/2017] [Accepted: 08/21/2017] [Indexed: 12/11/2022] Open
Abstract
Glucocorticoids (GC) are one of the most popular and versatile classes of drugs available to treat chronic inflammation and cancer, but side effects and resistance constrain their use. To overcome these hurdles, which are often related to the uniform tissue distribution of free GC and their short half-life in biological fluids, new delivery vehicles have been developed including PEGylated liposomes, polymeric micelles, polymer-drug conjugates, inorganic scaffolds, and hybrid nanoparticles. While each of these nanoformulations has individual drawbacks, they are often superior to free GC in many aspects including therapeutic efficacy when tested in cell culture or animal models. Successful application of nanomedicines has been demonstrated in various models of neuroinflammatory diseases, cancer, rheumatoid arthritis, and several other disorders. Moreover, investigations using human cells and first clinical trials raise the hope that the new delivery vehicles may have the potential to make GC therapies more tolerable, specific and efficient in the future.
Collapse
|
1230
|
Zhang X, Lin CC, Chan WKN, Liu KL, Yang ZJ, Zhang HQ. Augmented Anticancer Effects of Cantharidin with Liposomal Encapsulation: In Vitro and In Vivo Evaluation. Molecules 2017; 22:molecules22071052. [PMID: 28672816 PMCID: PMC6152072 DOI: 10.3390/molecules22071052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 01/10/2023] Open
Abstract
PEGylated liposomes have received much attention as pharmaceutical carriers to deliver chemotherapeutic agents for therapeutic purpose. The aim of this study was to prepare and characterize PEGylated liposome of cantharidin and investigate its therapeutic effect on human hepatocellular carcinoma treatment in vitro and in vivo. Liposomal cantharidin was evaluated for their anticancer effects in vitro using human hepatocellular carcinoma HepG2 cells and in vivo using HepG2-bearing nude mice compared to free drug. PEGylated liposome of cantharidin had a particle size of 129.9 nm and a high encapsulation efficacy of approximately 88.9%. The liposomal cantharidin had a higher anti-proliferative effect vis-à-vis free cantharidin in inducing G2/M cell cycle arrest and apoptosis. Liposomal cantharidin killed more HepG2 cancer cells at the same concentration equivalent to free cantharidin. Further study in vivo also showed that liposomal cantharidin achieved a higher tumor growth inhibition efficacy than free drug on hepatocellular carcinoma. As our study exhibited enhanced cytotoxicity against HepG2 cells and augmented tumor inhibitory effects in vivo, the results validate the potential value of cantharidin-liposome in improving the therapeutic efficacy of cantharidin for liver cancer.
Collapse
Affiliation(s)
- Xue Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Cong-Cong Lin
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | | | - Kang-Lun Liu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Zhi-Jun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Changshu Research Institute, Hong Kong Baptist University, Changshu Economic and Technological Development (CETD) Zone, Changshu 215500, China.
| | - Hong-Qi Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
1231
|
Yuan JJ, Qin FGF, Tu JL, Li B. Preparation, Characterization, and Antioxidant Activity Evaluation of Liposomes Containing Water-Soluble Hydroxytyrosol from Olive. Molecules 2017; 22:molecules22060870. [PMID: 28538693 PMCID: PMC6152771 DOI: 10.3390/molecules22060870] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 01/03/2023] Open
Abstract
Due to the multiple hydroxyl groups in its structure, hydroxytyrosol (HT) is very sensitive to air and light and has very strong instability and hydrophilicity that affect its biological activity. This study attempted to prepare liposomes containing water-soluble HT to improve the bioavailability and biocompatibility of the target drug. The preparation process factors (temperature, mass ratio of phospholipid (PL) and cholesterol (CH), Tween-80 volume, HT mass) were studied and response surface methodology (RSM) was applied to optimize the conditions. The results demonstrated that by using a temperature of 63 °C, mass ratio of PL and CH 4.5:1, HT mass 5 mg and Tween-80 volume of 6 mL, HT liposomes with an encapsulation efficiency (EE) of 45.08% were prepared. It was found that the particle sizes of the HT liposomes were well distributed in the range of 100–400 nm. Compared to free HT, prepared HT liposomes had better stability and a distinct slow release effect in vitro. Besides, HT liposomes presented better DPPH radical scavenging activity than free HT, which could be due to the fact that HT was encapsulated fully inside the liposomes. In addition, the encapsulation mechanism of HT was evaluated. In summary, the results indicated that HT liposome could enhance the antioxidant activity and was a promising formulation for prolonging the biological activity time of the target drug.
Collapse
Affiliation(s)
- Jiao-Jiao Yuan
- School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan 523808, China.
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Frank G F Qin
- School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan 523808, China.
| | - Jun-Ling Tu
- School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan 523808, China.
| | - Bing Li
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
1232
|
Pedoto A, Amar D. Liposomal Bupivacaine for Intercostal Nerve Block: Pricey or Priceless? Semin Thorac Cardiovasc Surg 2017; 29:538-539. [DOI: 10.1053/j.semtcvs.2017.08.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2017] [Indexed: 11/11/2022]
|