1
|
Cooper A, Subramaniam AB. Ultrahigh yields of giant vesicles obtained through mesophase evolution and breakup. SOFT MATTER 2024; 20:9547-9561. [PMID: 39618312 DOI: 10.1039/d4sm01109k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Self-assembly of dry amphiphilic lipid films on surfaces upon hydration is a crucial step in the formation of cell-like giant unilamellar vesicles (GUVs). GUVs are useful as biophysical models, as soft materials, as chassis for bottom-up synthetic biology, and in biomedical applications. Here via combined quantitative measurements of the molar yield and distributions of sizes and high-resolution imaging of the evolution of thin lipid films on surfaces, we report the discovery of a previously unknown pathway of lipid self-assembly which can lead to ultrahigh yields of GUVs of >50%. This yield is about 60% higher than any GUV yield reported to date. The "shear-induced fragmentation" pathway occurs in membranes containing 3 mol% of the poly(ethylene glycol) modified lipid PEG2000-DSPE (1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000]), when a lipid-dense foam-like mesophase forms upon hydration. The membranes in the mesophase fragment and close to form GUVs upon application of fluid shear. Experiments with varying mol% of PEG2000-DSPE and with lipids with partial molecular similarity to PEG2000-DSPE show that ultrahigh yields are only achievable under conditions where the lipid-dense mesophase forms. The increased yield of GUVs compared to mixtures without PEG2000-DSPE was general to flat supporting surfaces such as stainless steel sheets and to various lipid mixtures. In addition to increasing their accessibility as soft materials, these results demonstrate a route to obtaining ultrahigh yields of cell-sized liposomes using longstanding clinically-approved lipid formulations that could be useful for biomedical applications.
Collapse
Affiliation(s)
- Alexis Cooper
- Department of Chemistry and Biochemistry, University of California, Merced, CA 95343, USA
| | | |
Collapse
|
2
|
Raza M, Bharti H, Chauhan C, Singal A, Jha D, Ghosh PC, Nag A. Enhanced anti-malarial efficacy of mefloquine delivered via cationic liposome in a murine model of experimental cerebral malaria. Eur J Pharm Biopharm 2024; 197:114210. [PMID: 38340876 DOI: 10.1016/j.ejpb.2024.114210] [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: 08/12/2023] [Revised: 12/24/2023] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
Malaria is a longstanding global health challenge that continues to afflict over 90 countries located in tropical and subtropical regions of the globe. The rise of drug-resistant malarial parasites has curtailed the therapeutic efficacy of a number of once-effective anti-malarials, including mefloquine. In the present study, we have taken advantage of drug encapsulation approach to elevate the anti-malarial potential of mefloquine. Encouragingly, our findings unveil that liposomal formulations of mefloquine outperform equivalent doses of free mefloquine, both in laboratory cultures and in a murine model of malaria. Intriguingly, a cationic liposomal mefloquine formulation, administered at four successive doses of 3 mg/kg body weight, achieves complete resolution of cerebral malaria in the murine model while avoiding noticeable toxic repercussions. Altogether, our study furnishes pre-clinical validation for a therapeutic strategy that can remarkably enhance the drug efficacy, offering a revitalizing solution for failing anti-malarials.
Collapse
Affiliation(s)
- Mohsin Raza
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Hina Bharti
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Charu Chauhan
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Aakriti Singal
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Deepa Jha
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Prahlad C Ghosh
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India.
| |
Collapse
|
3
|
Makhlouf Z, Ali AA, Al-Sayah MH. Liposomes-Based Drug Delivery Systems of Anti-Biofilm Agents to Combat Bacterial Biofilm Formation. Antibiotics (Basel) 2023; 12:antibiotics12050875. [PMID: 37237778 DOI: 10.3390/antibiotics12050875] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
All currently approved antibiotics are being met by some degree of resistance by the bacteria they target. Biofilm formation is one of the crucial enablers of bacterial resistance, making it an important bacterial process to target for overcoming antibiotic resistance. Accordingly, several drug delivery systems that target biofilm formation have been developed. One of these systems is based on lipid-based nanocarriers (liposomes), which have shown strong efficacy against biofilms of bacterial pathogens. Liposomes come in various types, namely conventional (charged or neutral), stimuli-responsive, deformable, targeted, and stealth. This paper reviews studies employing liposomal formulations against biofilms of medically salient gram-negative and gram-positive bacterial species reported recently. When it comes to gram-negative species, liposomal formulations of various types were reported to be efficacious against Pseudomonas aeruginosa, Escherichia coli, Acinetobacter baumannii, and members of the genera Klebsiella, Salmonella, Aeromonas, Serratia, Porphyromonas, and Prevotella. A range of liposomal formulations were also effective against gram-positive biofilms, including mostly biofilms of Staphylococcal strains, namely Staphylococcus aureus, Staphylococcus epidermidis, and Staphylococcus saprophyticus subspecies bovis, followed by Streptococcal strains (pneumonia, oralis, and mutans), Cutibacterium acnes, Bacillus subtilis, Mycobacterium avium, Mycobacterium avium subsp. hominissuis, Mycobacterium abscessus, and Listeria monocytogenes biofilms. This review outlines the benefits and limitations of using liposomal formulations as means to combat different multidrug-resistant bacteria, urging the investigation of the effects of bacterial gram-stain on liposomal efficiency and the inclusion of pathogenic bacterial strains previously unstudied.
Collapse
Affiliation(s)
- Zinb Makhlouf
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Amaal Abdulraqeb Ali
- Biomedical Engineering Program, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Mohammad Hussein Al-Sayah
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
4
|
Fan CH, Wu N, Yeh CK. Enhanced sonodynamic therapy by carbon dots-shelled microbubbles with focused ultrasound. ULTRASONICS SONOCHEMISTRY 2023; 94:106342. [PMID: 36842213 PMCID: PMC9988694 DOI: 10.1016/j.ultsonch.2023.106342] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/07/2023] [Accepted: 02/20/2023] [Indexed: 05/28/2023]
Abstract
Sonodynamic therapy involving the non-invasive and local generation of lethal reactive oxygen species (ROS) via ultrasound (US) with sonosensitizers has been proposed as an emerging tumor therapy strategy. However, such therapy is usually associated with inertial cavitation and unnecessary damage to healthy tissue because current sonosensitizers have insufficient sensitivity to US. Here, we report the use of a new proposed sonosensitizer, carbon dots (C-dots), to assemble microbubbles with a gas core (C-dots MBs). As the C-dots were directly integrated into the MB shell, they could effectively absorb the energy of inertial cavitation and transfer it to ROS. Our results revealed the appearance of 1O2, •OH, and H2O2 after US irradiation of C-dots MBs. In in vitro experiments, treatment with C-dots MBs plus US induced lipid peroxidation, elevation of intracellular ROS, and apoptosis in 32.5%, 45.3%, and 50.1% of cells respectively. In an animal solid tumor model, treatment with C-dots MBs plus US resulted in a 3-fold and 2.5-fold increase in the proportion of ROS-damaged cells and apoptotic cells, respectively, compared to C-dots MBs alone. These results will pave the way for the design of novel multifunctional sonosensitizers for SDT tumor therapy.
Collapse
Affiliation(s)
- Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Nan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
5
|
Suzuki H, Adler A, Huang T, Kuramochi A, Ohba Y, Sato Y, Nakamura N, Manivel VA, Ekdahl KN, Nilsson B, Ishihara K, Teramura Y. Impact of spontaneous liposome modification with phospholipid polymer-lipid conjugates on protein interactions. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2022; 23:845-857. [PMID: 36518982 PMCID: PMC9744213 DOI: 10.1080/14686996.2022.2146466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 06/17/2023]
Abstract
Liposome surface coating has been studied to avoid the immunological responses caused by the complement system, and alternative materials to poly(ethylene glycol) (PEG) have been explored recently since the production of anti-PEG IgM antibodies has been found in humans. We previously reported a liposome coating with poly(2-methacryloyloxyethyl phosphorylcholine) (poly(MPC))-conjugated lipids (PMPC-lipids) and demonstrated its protective effect on blood protein interactions. Here, we attempted to modify the liposome surface by exogenously adding PMPC-lipids, which were spontaneously incorporated into the outer membrane via hydrophobic interactions. The polymerization degree of the PMPC segment was regulated from 10 to 100. The incorporated ratio of PMPC-lipid increased with a decrease in the degree of PMPC polymerization. Due to surface modification with PMPC-lipids, increase in the length of the PMPC-chain increased the size of the liposomes. The modified liposomes were kept stable for 14 d in terms of their size, polydispersity, and surface properties, where approximately 70% of PMPC-lipids were incorporated into the liposome surface. We demonstrated that liposome surface modification with PMPC-lipids can inhibit protein adsorption when exposed to serum, regardless of the degree of polymerization of PMPC. In addition, the PMPC-lipid modified surface was not recognized by the anti-PEG IgM antibody, whereas PEG-lipid was recognized by the antibody. Thus, we successfully fabricated an inert liposome surface via spontaneous modification with PMPC-lipids, where only the outer bilayer surface was modified. This technique can be available for full loading of water-soluble active pharmaceutical ingredient inside the modified liposome.
Collapse
Affiliation(s)
- Haruna Suzuki
- Department of Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| | - Anna Adler
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
| | - Tianwei Huang
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Akiko Kuramochi
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | - Yoshiro Ohba
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | - Yuya Sato
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Naoko Nakamura
- Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| | - Vivek Anand Manivel
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
| | - Kristina N Ekdahl
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
- Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
| | - Kazuhiko Ishihara
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Yuji Teramura
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
- Master’s/Doctoral Program in Life Science Innovation (T-LSI), University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
6
|
Saad MA, Hasan T. Spotlight on Photoactivatable Liposomes beyond Drug Delivery: An Enabler of Multitargeting of Molecular Pathways. Bioconjug Chem 2022; 33:2041-2064. [PMID: 36197738 DOI: 10.1021/acs.bioconjchem.2c00376] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The potential of photoactivating certain molecules, photosensitizers (PS), resulting in photochemical processes, has long been realized in the form of photodynamic therapy (PDT) for the management of several cancerous and noncancerous pathologies. With an improved understanding of the photoactivation process and its broader implications, efforts are being made to exploit the various facets of photoactivation, PDT, and the associated phenomenon of photodynamic priming in enhancing treatment outcomes, specifically in cancer therapeutics. The parallel emergence of nanomedicine, specifically liposome-based nanoformulations, and the convergence of the two fields of liposome-based drug delivery and PDT have led to the development of unique hybrid systems, which combine the exciting features of liposomes with adequate complementation through the photoactivation process. While initially liposomes carrying photosensitizers (PSs) were developed for enhancing the pharmacokinetics and the general applicability of PSs, more recently, PS-loaded liposomes, apart from their utility in PDT, have found several applications including enhanced targeting of drugs, coloading multiple therapeutic agents to enhance synergistic effects, imaging, priming, triggering drug release, and facilitating the escape of therapeutic agents from the endolysosomal complex. This review discusses the design strategies, potential, and unique attributes of these hybrid systems, with not only photoactivation as an attribute but also the ability to encapsulate multiple agents for imaging, biomodulation, priming, and therapy referred to as photoactivatable multiagent/inhibitor liposomes (PMILS) and their targeted versions─targeted PMILS (TPMILS). While liposomes have formed their own niche in nanotechnology and nanomedicine with several clinically approved formulations, we try to highlight how using PS-loaded liposomes could address some of the limitations and concerns usually associated with liposomes to overcome them and enhance their preclinical and clinical utility in the future.
Collapse
Affiliation(s)
- Mohammad A Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States.,Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
7
|
Ishima Y, Yamazaki N, Chuang VTG, Shimizu T, Ando H, Ishida T. A Maleimide-Terminally Modified PEGylated Liposome Induced the Accelerated Blood Clearance Independent of the Production of Anti-PEG IgM Antibodies. Biol Pharm Bull 2022; 45:1518-1524. [PMID: 36184510 DOI: 10.1248/bpb.b22-00389] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
PEGylated liposomes (PL) lose their long-circulating characteristic when administered repeatedly, called the accelerated blood clearance (ABC) phenomenon. The ABC phenomenon is generally thought to occur when the anti-polyethylene glycol (PEG) antibody (anti-PEG immunoglobulin M (IgM)) expressed in the spleen B cells triggered by the first dose of PL binds to the second and subsequent doses of PL, leading to activation of the complement system. MAL-PEG-DSPE, a PEG lipid with a maleimide (MAL) group at the PEG terminal, is used in various studies as a linker for ligand-bound liposomes such as antibody-modified liposomes. However, most ABC phenomenon research used PL with a terminal methoxy group (PL-OCH3). In this study, we prepared MAL-PEG-DSPE liposomes (PL-MAL) to evaluate the effect of PL-MAL on the ABC phenomenon induction compared to PL-OCH3. Pharmacokinetic, anti-PEG IgM secretion and complement activation analyses of these liposomes were conducted in mice. Interestingly, despite C3 bound to the surface of the initially administered PL-MAL, the administered PL-MAL showed high blood retention, demonstrating the same results as PL-OCH3. On the other hand, although the secretion of anti-PEG IgM induced by PL-MAL was lower than PL-OCH3, the second dose of PL-MAL rapidly disappeared from the blood. These results suggest that the antibody produced from the first dose of PL-MAL binds to the second dose of PL-MAL, thereby activating C3 to act as an opsonin which promotes phagocytic uptake. In conclusion, PL-MAL induced the ABC phenomenon independent of the production of IgM antibodies against PEG. This study provides valuable findings for further studies using ligand-bound liposomes.
Collapse
Affiliation(s)
- Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Nio Yamazaki
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Victor T G Chuang
- Curtin Medical School, Faculty of Health Sciences, Curtin University
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| |
Collapse
|
8
|
Sakurai Y. Development of siRNA Delivery System by Lipid Nanoparticles Modified with Functional Materials for Cancer Treatment. Biol Pharm Bull 2022; 45:972-977. [DOI: 10.1248/bpb.b22-00317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yu Sakurai
- Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
9
|
Lee H, Moon H, Kim HR. Effects of Lipid Shape and Interactions on the Conformation, Dynamics, and Curvature of Ultrasound-Responsive Liposomes. Pharmaceutics 2022; 14:1512. [PMID: 35890407 PMCID: PMC9320727 DOI: 10.3390/pharmaceutics14071512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/10/2022] [Accepted: 07/19/2022] [Indexed: 12/10/2022] Open
Abstract
We perform coarse-grained molecular dynamics simulations of bilayers composed of various lipids and cholesterol at their different ratios. Simulations show that cholesterol-lipid interactions restrict the lateral dynamics of bilayers but also promote bilayer curvature, indicating that these opposite effects simultaneously occur and thus cannot significantly influence bilayer stability. In contrast, lyso-lipids effectively pack the vacancy in the bilayer composed of cone-shaped lipids and thus reduce bilayer dynamics and curvature, showing that bilayers are more significantly stabilized by lyso-lipids than by cholesterol, in agreement with experiments. In particular, the bilayer composed of cone-shaped lipids shows higher dynamics and curvature than does the bilayer composed of cylindrical-shaped lipids. To mimic ultrasound, a high external pressure was applied in the direction of bilayer normal, showing the formation of small pores that are surrounded by hydrophilic lipid headgroups, which can allow the release of drug molecules encapsulated into the liposome. These findings help to explain experimental observations regarding that liposomes are more significantly stabilized by lyso-lipids than by cholesterol, and that the liposome with cone-shaped lipids more effectively releases drug molecules upon applying ultrasound than does the liposome with cylindrical-shaped lipids.
Collapse
Affiliation(s)
- Hwankyu Lee
- Department of Chemical Engineering, Dankook University, Yongin-si 16890, Korea
| | - Hyungwon Moon
- R&D Center, IMGT Co., Ltd., 172, Dolma-ro, Bundang-gu, Seongnam-si 13605, Korea;
| | - Hyun-Ryoung Kim
- R&D Center, IMGT Co., Ltd., 172, Dolma-ro, Bundang-gu, Seongnam-si 13605, Korea;
| |
Collapse
|
10
|
Sato W, Zajkowski T, Moser F, Adamala KP. Synthetic cells in biomedical applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1761. [PMID: 34725945 PMCID: PMC8918002 DOI: 10.1002/wnan.1761] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
Synthetic cells are engineered vesicles that can mimic one or more salient features of life. These features include directed localization, sense-and-respond behavior, gene expression, metabolism, and high stability. In nanomedicine, many of these features are desirable capabilities of drug delivery vehicles but are difficult to engineer. In this focus article, we discuss where synthetic cells offer unique advantages over nanoparticle and living cell therapies. We review progress in the engineering of the above life-like behaviors and how they are deployed in nanomedicine. Finally, we assess key challenges synthetic cells face before being deployed as drugs and suggest ways to overcome these challenges. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Wakana Sato
- 1 Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN US
| | - Tomasz Zajkowski
- Centre of New Technologies, University of Warsaw, S. Banacha 2c, 02-097 Warsaw, Poland
- USRA at NASA Ames Research Center, Mountain View, CA 94035
- Blue Marble Space Institute of Science, 600 1st Avenue, Seattle WA 98104
| | - Felix Moser
- Synlife, Inc., One Kendall Square Suite B4401, Cambridge, MA 20139
| | - Katarzyna P. Adamala
- 1 Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN US
| |
Collapse
|
11
|
Abu-Dief AM, Alsehli M, Awaad A. A higher dose of PEGylated gold nanoparticles reduces the accelerated blood clearance phenomenon effect and induces spleen B lymphocytes in albino mice. Histochem Cell Biol 2022; 157:641-656. [PMID: 35157114 DOI: 10.1007/s00418-022-02086-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2022] [Indexed: 11/30/2022]
Abstract
Polyethylene glycol (PEG) is a multifunctional polymer that has many uses in medical and biological applications. Recently, PEG has been mainly used in developing nanomaterial-based drug delivery systems (DDS). PEG is characterized by its high solubility, biological inertness, and ability to escape from immune cells (stealthiness) after systemic injection. The most challenging problem for PEGylated nanomaterials is their rapid elimination from the bloodstream after repeated doses of systemic injection, called accelerated blood clearance (ABC). Therefore, in this study, the effect of PEGylated nanomaterial dose concentration on ABC induction will be investigated using quantitative, histological, and immunohistochemical analyses. A higher dose concentration (2 mg/kg) of PEGylated gold nanoparticles (PEG-coated AuNPs) reduced the ABC phenomenon when intravenously injected into mice preinjected with the same dose. In contrast, a lower dose concentration (< 1 mg/kg) significantly induced the ABC phenomenon by the rapid elimination of the second dose of PEG-coated AuNPs from the bloodstream. To explain the relationship between the dose concentration (from PEG and AuNPs) and the induction of ABC, the biodistribution of PEG-coated AuNPs in liver and spleen [reticuloendothelial systems (RES)-rich organs] was investigated. The injected dose of PEG-coated AuNPs accumulated mainly in the hepatic Kupffer cells and hepatocytes. Similarly, spleen red pulp received a higher amount of the injected dose of PEG-coated AuNPs. However, the biodistriution profiles of PEG-coated AuNPs after the first and second dose for different dose concentrations varied in RES-rich organs. Additionally, the number of B lymphocytes, which have an important role in producing anti-PEG immunoglobulin (Ig)M, was affected by the repeated dose of PEG-coated AuNPs in the spleen. Therefore, for effective nanomaterial-based DDS development, dose optimization of PEG molecules that express PEGylated nanomaterials is important to reduce the ABC phenomenon effect. The ideal concentration of PEG molecules used to coat nanomaterials and the role of RES-rich organs must be determined to control the ABC phenomenon effect of PEGylated nanomaterials.
Collapse
Affiliation(s)
- Ahmed M Abu-Dief
- Department of Chemistry, College of Science, Taibah University, P.O. Box 344, Madinah, Saudi Arabia.,Department of Chemistry, Faculty of Science, Sohag University, Sohag, 82524, Egypt
| | - Mosa Alsehli
- Department of Chemistry, College of Science, Taibah University, P.O. Box 344, Madinah, Saudi Arabia
| | - Aziz Awaad
- Department of Zoology, Faculty of Science, Sohag University, Sohag, 52824, Egypt.
| |
Collapse
|
12
|
Chen M, Sheu MT, Cheng TL, Roffler SR, Lin SY, Chen YJ, Cheng YA, Cheng JJ, Chang HY, Wu TY, Kao AP, Ho YS, Chuang KH. A novel anti-tumor/anti-tumor-associated fibroblast/anti-mPEG tri-specific antibody to maximize the efficacy of mPEGylated nanomedicines against fibroblast-rich solid tumor. Biomater Sci 2021; 10:202-215. [PMID: 34826322 DOI: 10.1039/d1bm01218e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The therapeutic efficacy of methoxypolyethylene glycol (mPEG)-coated nanomedicines in solid tumor treatment is hindered by tumor-associated fibroblasts (TAFs), which promote tumor progression and form physical barriers. We developed an anti-HER2/anti-FAP/anti-mPEG tri-specific antibody (TsAb) for one-step conversion of mPEG-coated liposomal doxorubicin (Lipo-Dox) to immunoliposomes, which simultaneously target HER2+ breast cancer cells and FAP+ TAFs. The non-covalent modification did not adversely alter the physical characteristics and stability of Lipo-Dox. The TsAb-Lipo-Dox exhibited specific targeting and enhanced cytotoxicity against mono- and co-cultured HER2+ breast cancer cells and FAP+ TAFs, compared to bi-specific antibody (BsAb) modified or unmodified Lipo-Dox. An in vivo model of human breast tumor containing TAFs also revealed the improved tumor accumulation and therapeutic efficacy of TsAb-modified mPEGylated liposomes without signs of toxicity. Our data indicate that arming clinical mPEGylated nanomedicines with the TsAb is a feasible and applicable approach for overcoming the difficulties caused by TAFs in solid tumor treatment.
Collapse
Affiliation(s)
- Michael Chen
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Ming-Thau Sheu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Tian-Lu Cheng
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shyr-Yi Lin
- Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei, Taiwan.,Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jou Chen
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Yi-An Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Jing-Jy Cheng
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
| | - Hsin-Yu Chang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Tung-Yun Wu
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan
| | - An-Pei Kao
- Stemforce Biotechnology Co., Ltd, Chiayi City, Taiwan
| | - Yuan-Soon Ho
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.,School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan. .,Cancer Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan.,Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei, Taiwan. .,Ph.D Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan.,Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
13
|
Petrini M, Lokerse WJM, Mach A, Hossann M, Merkel OM, Lindner LH. Effects of Surface Charge, PEGylation and Functionalization with Dipalmitoylphosphatidyldiglycerol on Liposome-Cell Interactions and Local Drug Delivery to Solid Tumors via Thermosensitive Liposomes. Int J Nanomedicine 2021; 16:4045-4061. [PMID: 34163158 PMCID: PMC8214027 DOI: 10.2147/ijn.s305106] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/24/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose Previous studies demonstrated the possibility of targeting tumor-angiogenic endothelial cells with positively charged nanocarriers, such as cationic liposomes. We investigated the active targeting potential of positively charged nanoparticles in combination with the heat-induced drug release function of thermosensitive liposomes (TSL). This novel dual-targeted approach via cationic TSL (CTSL) was thoroughly explored using either a novel synthetic phospholipid 1,2-dipalmitoyl-sn-glycero-3-phosphodiglycerol (DPPG2) or a conventional polyethylene glycol (PEG) surface modification. Anionic particles containing either DPPG2 or PEG were also included in the study to highlight difference in tumor enrichment driven by surface charge. With this study, we aim to provide a deep insight into the main differences between DPPG2- and PEG-functionalized liposomes, focusing on the delivery of a well-known cytotoxic drug (doxorubicin; DOX) in combination with local hyperthermia (HT, 41–43°C). Materials and Methods DPPG2- and PEG-based cationic TSLs (PG2-CTSL/PEG-CTSL) were thoroughly analyzed for size, surface charge, and heat-triggered DOX release. Cancer cell targeting and DOX delivery was evaluated by FACS, fluorescence imaging, and HPLC. In vivo particle behavior was analyzed by assessing DOX biodistribution with local HT application in tumor-bearing animals. Results The absence of PEG in PG2-CTSL promoted more efficient liposome–cell interactions, resulting in a higher DOX delivery and cancer cell toxicity compared with PEG-CTSL. By exploiting the dual-targeting function of CTSLs, we were able to selectively trigger DOX release in the intracellular compartment by HT. When tested in vivo, local HT promoted an increase in intratumoral DOX levels for all (C)TSLs tested, with DOX enrichment factors ranging from 3 to 14-fold depending on the type of formulation. Conclusion Cationic particles showed lower hemocompatibility than their anionic counterparts, which was partially mitigated when PEG was grafted on the liposome surface. DPPG2-based anionic TSL showed optimal local drug delivery compared to all other formulations tested, demonstrating the potential advantages of using DPPG2 lipid in designing liposomes for tumor-targeted applications.
Collapse
Affiliation(s)
- Matteo Petrini
- Department of Internal Medicine III, University Hospital, Ludwig Maximilian University, Munich, Germany.,Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig Maximilian University, Munich, Germany
| | - Wouter J M Lokerse
- Department of Internal Medicine III, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Agnieszka Mach
- Department of Internal Medicine III, University Hospital, Ludwig Maximilian University, Munich, Germany
| | | | - Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig Maximilian University, Munich, Germany
| | - Lars H Lindner
- Department of Internal Medicine III, University Hospital, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
14
|
Cheng WJ, Lin SY, Chen M, Chen LC, Ho HO, Chuang KH, Sheu MT. Active Tumoral/Tumor Environmental Dual-Targeting by Non-Covalently Arming with Trispecific Antibodies or Dual-Bispecific Antibodies on Docetaxel-Loaded mPEGylated Nanocarriers to Enhance Chemotherapeutic Efficacy and Minimize Systemic Toxicity. Int J Nanomedicine 2021; 16:4017-4030. [PMID: 34140769 PMCID: PMC8203191 DOI: 10.2147/ijn.s301237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE This study was aimed at developing the trispecific antibodies (anti-EGFR/anti-FAP/anti-mPEG, TsAb) or dual bispecific antibodies (anti-EGFR/anti-mPEG and anti-FAP/anti-mPEG) docetaxel (DTX)-loaded mPEGylated lecithin-stabilized micelles (mPEG-lsbPMs) for improving the targeting efficiency and therapeutic efficacy. METHODS mPEG-lsbPMs were simply prepared via thin film method. The trispecific antibodies or bispecific antibodies bound the mPEG-lsbPMs by anti-mPEG Fab fragment. The formulations were characterized by DLS and TEM; in vitro and in vivo studies were also conducted to evaluate the cellular uptake, cell cytotoxicity and therapeutic efficacy. RESULTS The particle sizes of mPEG-lsbPMs with or without the antibodies were around 100 nm; the formulations showed high encapsulation efficiencies of 97.12%. The TsAb and dual bispecific antibodies were fabricated and demonstrated their targeting ability. Two EGFR-overexpressed cell lines (HT-29 and MIA PaCa-2) were co-cultured with FAP-overexpressed WS1 cells (HT-29/WS1; MIA PaCa-2/WS1) to mimic a tumor coexisting in the tumor microenvironment. Cellular binding study revealed that the binding of anti-FAP micelles to three co-culture ratios (4:1, 1:1, and 1:4) of HT-29/EGFR to WS1/FAP was significantly higher than that for TsAb micelles and dual (1:1) micelles, and the binding of those targeting antibodies to WS1/FAP and MIA PaCa-2/EGFR was equally efficacious resulting in a similar binding amount of the TsAb and dual BsAbs (1:1) with the co-culture of MIA PaCa-2/EGFR and WS1/FAP at a 1:1 ratio. Antitumor efficacy study showed that treatment with DTX-loaded mPEG-lsbPMs modified with or without BsAbs, dual BsAbs (1:1), and TsAbs was enhanced in inhibiting tumor growth compared with that for Tynen® while showing fewer signs of adverse effects. CONCLUSION Active targeting of both tumors and TAF-specific antigens was able to increase the affinity of DTX-loaded mPEG-lsbPMs toward tumor cells and TAFs leading to successive uptake by tumor cells or TAFs which enhanced their chemotherapeutic efficacy against antigen-positive cancer cells.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/administration & dosage
- Antibodies, Bispecific/chemistry
- Antibodies, Bispecific/pharmacology
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/pharmacology
- Cancer-Associated Fibroblasts/drug effects
- Cell Line, Tumor
- Coculture Techniques
- Docetaxel/administration & dosage
- Docetaxel/pharmacokinetics
- Drug Carriers/administration & dosage
- Drug Carriers/chemistry
- Drug Delivery Systems/methods
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/immunology
- Humans
- Injections, Intradermal
- Lecithins/chemistry
- Male
- Mice, Nude
- Micelles
- Particle Size
- Polyethylene Glycols/chemistry
- Rats, Sprague-Dawley
- Tumor Microenvironment/drug effects
- Xenograft Model Antitumor Assays
- Mice
- Rats
Collapse
Affiliation(s)
- Wei-Jie Cheng
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Shyr-Yi Lin
- Division of Gastroenterology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Michael Chen
- PhD Program in Clinical Drug Development of Chinese Herbal Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ling-Chun Chen
- Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Hsiu-O Ho
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Hsiang Chuang
- PhD Program in Clinical Drug Development of Chinese Herbal Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Ming-Thau Sheu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
15
|
Deodhar S, Dash AK, North EJ, Hulce M. Development and In Vitro Evaluation of Long Circulating Liposomes for Targeted Delivery of Gemcitabine and Irinotecan in Pancreatic Ductal Adenocarcinoma. AAPS PharmSciTech 2020; 21:231. [PMID: 32778980 DOI: 10.1208/s12249-020-01745-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/05/2020] [Indexed: 12/18/2022] Open
Abstract
The classically used nontargeted chemotherapeutic approach to pancreatic cancer has a dual drawback of suboptimal drug delivery at the target site and the systemic side effects produced by the unfettered exposure of the drug to healthy tissue. This study has the objective of developing novel poly(2-ethyl-2-oxazoline) (PETOX)-based long circulating liposomes loaded with gemcitabine and irinotecan for the treatment of pancreatic ductal adenocarcinoma, with a juxtaposition to PEGylated and uncoated liposomes. A PETOX-cholesteryl chloroformate lipopolymer conjugate (PETOX-ChC) with a carbonate linkage was prepared and characterized by 1H NMR, FTIR, and DSC. Liposomes were prepared using the thin film hydration technique followed by freeze-thaw and membrane extrusion methods. Liposome characterization includes particle size determination, zeta potential determination using a zetameter, and structural elucidation using 31P NMR and cryo-TEM. The PETOXylated liposomes showed a particle size of 180.1 ± 2.2 nm and a zeta potential of - 33.63 ± 1.23 mV. The liposomal combination therapy of gemcitabine and irinotecan was found to have an IC50 value 39 times lower in comparison to the drug combination in solution, while the PEGylated and PETOXylated liposomes showed IC50 values 1.6 times lower and 2 times lower than that of uncoated liposomes, respectively, against Mia PaCa II pancreatic cancer cell line. The PEGylated and PETOXylated liposomes showed 4.1 and 5.4 times slower macrophagial uptake in vitro in comparison to the uncoated liposomes respectively. The PEGylated liposomes showed 11% higher in vitro macrophagial uptake in comparison to PETOXylated liposomes.
Collapse
|
16
|
Abstract
The aim of this study was to evaluate the characterized hydration method to prepare nanoparticles using Soluplus, a block copolymer with amphipathic properties, and distearoyl phosphatidyl ethanolamine (DSPE)-PEG2000 owing to particle size distribution, zeta potential, particle stability, and transmission electron microscopy (TEM) observed and 31P-NMR spectra. The results showed that, in a suspension of DSPE-PEG2000 and Soluplus at a ratio of 1/1, the prepared microparticles were stable for five days in the dark and at 25 °C. It was also confirmed that the 1/1 suspension of DSPE-PEG2000/Soluplus was stable for five days under the same conditions with the magnesium chloride solution. TEM measurements confirmed the presence of micelle-like particles of 50 to 150 nm in the 1/1 ratio mix of DSPE-PEG2000/Soluplus. 31P-NMR spectral data confirmed that DPSE-PEG2000/Soluplus at mixing ratio of 1/1 has a strong intermolecular with the phosphate group, indicated by the fact that the peak shift and the full width at half maximum were the largest compared with DSPE-PEG2000 with the intermolecular interaction. On the basis of the findings of this study, we conclude that microparticles can be formed using DSPE-PEG2000 and Soluplus via the hydration method, and that the optimum weight ratio of DSPE-PEG2000 to Soluplus is 1/1.
Collapse
|
17
|
Shinde G, Desai P, Shelke S, Patel R, Bangale G, Kulkarni D. Mometasone furoate-loaded aspasomal gel for topical treatment of psoriasis: formulation, optimization, in vitro and in vivo performance. J DERMATOL TREAT 2020; 33:885-896. [PMID: 32603203 DOI: 10.1080/09546634.2020.1789043] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Present investigation was aimed to develop aspasomal gel of Mometasone Furoate for the treatment of Psoriasis that are biologically active and deliver drug at controlled rate and decrease dosing frequency. METHODS The vesicles were fabricated using film hydration method and optimized using 32 factorial Design. Prepared formulations were evaluated for percent drug loading, vesicle size, Zeta potential, polydispersity index and morphological studies. Gel was prepared using carbopol by loading optimized drug loaded asposomes and was evaluated for drug content, pH, viscosity and spreadability. The drug release study from the gel was done using dialysis membrane and goat skin. Anti- oxidant potency of the prepared aspasomal gel was determined by Ferric Reducing Assay whereas, in-vivo performance for inflammation and skin irritation was carried out using Wistar rats. RESULTS Optimized aspasomes demonstrated desired properties for entrapment efficiency (74.72 ± 1.8), vesicle size (282.9 ± 1.7), polydispersity index (0.2), zeta potential (-20.2 mV) with spherical shape. The results recorded for drug release from the optimized aspasomal gel exhibited sustained release (24h) compared to the marketed cream (5h). Depot formation of Mometasone furoate loaded aspasomal gel in the epidermis was confirmed by ex vivo skin penetration study by using fluorescent marker. In-vivo study revealed no any irritation and inflammation to the skin promoting drug delivery system to treat psoriasis. CONCLUSION In conclusion, Mometasone furoate loaded aspasomal gel releases the drug for longer duration of time and reduce dosing frequency, providing the new dimension for the treatment of psoriasis.
Collapse
Affiliation(s)
- Gajanan Shinde
- Department of Pharmaceutics, Parul Institute of Pharmacy, Faculty of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Pankhita Desai
- Department of Pharmaceutics, Parul Institute of Pharmacy, Faculty of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Santosh Shelke
- Department of Pharmaceutics, Srinath College of Pharmacy, Aurangabad, Maharashtra, India
| | - Rakesh Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Faculty of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Ganesh Bangale
- Department of Pharmaceutics, Government College of Amravati, Amravati, Maharashtra, India
| | - Deepak Kulkarni
- Department of Pharmaceutics, Srinath College of Pharmacy, Aurangabad, Maharashtra, India
| |
Collapse
|
18
|
In Vivo Assessment of Thermosensitive Liposomes for the Treatment of Port Wine Stains by Antifibrinolytic Site-Specific Pharmaco-Laser Therapy. Pharmaceutics 2020; 12:pharmaceutics12060591. [PMID: 32630457 PMCID: PMC7356038 DOI: 10.3390/pharmaceutics12060591] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/13/2020] [Accepted: 06/22/2020] [Indexed: 01/10/2023] Open
Abstract
Antifibrinolytic site-specific pharmaco-laser therapy (SSPLT) is an experimental treatment modality for refractory port wine stains (PWS). Conceptually, antifibrinolytic drugs encapsulated in thermosensitive liposomes are delivered to thrombi that form in semi-photocoagulated PWS blood vessels after conventional laser treatment. Local release of antifibrinolytics is induced by mild hyperthermia, resulting in hyperthrombosis and complete occlusion of the target blood vessel (clinical endpoint). In this study, 20 thermosensitive liposomal formulations containing tranexamic acid (TA) were assayed for physicochemical properties, TA:lipid ratio, encapsulation efficiency, and endovesicular TA concentration. Two candidate formulations (DPPC:DSPE-PEG, DPPC:MPPC:DSPE-PEG) were selected based on optimal properties and analyzed for heat-induced TA release at body temperature (T), phase transition temperature (Tm), and at T > Tm. The effect of plasma on liposomal stability at 37 °C was determined, and the association of liposomes with platelets was examined by flow cytometry. The accumulation of PEGylated phosphocholine liposomes in laser-induced thrombi was investigated in a hamster dorsal skinfold model and intravital fluorescence microscopy. Both formulations did not release TA at 37 °C. Near-complete TA release was achieved at Tm within 2.0–2.5 min of heating, which was accelerated at T > Tm. Plasma exerted a stabilizing effect on both formulations. Liposomes showed mild association with platelets. Despite positive in vitro results, fluorescently labeled liposomes did not sufficiently accumulate in laser-induced thrombi in hamsters to warrant their use in antifibrinolytic SSPLT, which can be solved by coupling thrombus-targeting ligands to the liposomes.
Collapse
|
19
|
Lee H. Molecular Simulations of PEGylated Biomolecules, Liposomes, and Nanoparticles for Drug Delivery Applications. Pharmaceutics 2020; 12:E533. [PMID: 32531886 PMCID: PMC7355693 DOI: 10.3390/pharmaceutics12060533] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/17/2022] Open
Abstract
Since the first polyethylene glycol (PEG)ylated protein was approved by the FDA in 1990, PEGylation has been successfully applied to develop drug delivery systems through experiments, but these experimental results are not always easy to interpret at the atomic level because of the limited resolution of experimental techniques. To determine the optimal size, structure, and density of PEG for drug delivery, the structure and dynamics of PEGylated drug carriers need to be understood close to the atomic scale, as can be done using molecular dynamics simulations, assuming that these simulations can be validated by successful comparisons to experiments. Starting with the development of all-atom and coarse-grained PEG models in 1990s, PEGylated drug carriers have been widely simulated. In particular, recent advances in computer performance and simulation methodologies have allowed for molecular simulations of large complexes of PEGylated drug carriers interacting with other molecules such as anticancer drugs, plasma proteins, membranes, and receptors, which makes it possible to interpret experimental observations at a nearly atomistic resolution, as well as help in the rational design of drug delivery systems for applications in nanomedicine. Here, simulation studies on the following PEGylated drug topics will be reviewed: proteins and peptides, liposomes, and nanoparticles such as dendrimers and carbon nanotubes.
Collapse
Affiliation(s)
- Hwankyu Lee
- Department of Chemical Engineering, Dankook University, Yongin 16890, Korea
| |
Collapse
|
20
|
Zhao Y, Cai F, Shen X, Su H. A high stable pH-temperature dual-sensitive liposome for tuning anticancer drug release. Synth Syst Biotechnol 2020; 5:103-110. [PMID: 32596520 PMCID: PMC7301211 DOI: 10.1016/j.synbio.2020.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/03/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
In order to improve the targeting and availability of liposomes to cancer cells, the temperature sensitivity of 1, 2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and the pH sensitivity of PASP in PASP-g-C8 are incorporated in a drug delivery system. A composite pH-temperature dual-sensitive liposomes (CPTLPs) was obtained as an efficient drug delivery system. The bionic bilayer is self-assembled by cholesterol/cationic temperature-sensitive lipids as base layer and pH-sensitive octylamine grafted poly aspartic acid (PASP-g-C8) as anchors coated outside. Cytarabine (CYT) was chosen as a model drug. SEM and DLS were used to observe the morphology characteristics of CPTLPs in different micro environment. The results demonstrated that the CPTLPs remained active in both normal (pH7.4 and 37 °C) and tumor tissues (pH 5.0 and 42 °C). As a stable colloidal system, the zeta potential of CPTSLs was -41.6 mV. In vitro drug-release experiments, the CTY encapsulated dual-sensitive liposomes, CPTSLs(+), not only have significant pH-temperature sensitivity but have more prolonged release in vitro than control groups. MTT tests results indicated that the cell apoptotic effects induced by CPTSLs(+) were nearly 30% higher than the naked drug CTY in HepG2 cells, and 20% lower apoptotic in vero cells. The CPTSLs(+) sustained a stable emulsion form, less toxic effects on normal cells, and exhibited a good pH-temperature sensitivity, thus expected to be a promising tumor targeting drug delivery.
Collapse
Affiliation(s)
- Yilin Zhao
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology(BUCT), 15 BeiSanhuan East Road, ChaoYang District, Beijing, 100029, PR China
| | - Fuli Cai
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology(BUCT), 15 BeiSanhuan East Road, ChaoYang District, Beijing, 100029, PR China
| | - Xiangyi Shen
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology(BUCT), 15 BeiSanhuan East Road, ChaoYang District, Beijing, 100029, PR China
| | - Haijia Su
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology(BUCT), 15 BeiSanhuan East Road, ChaoYang District, Beijing, 100029, PR China
| |
Collapse
|
21
|
Wen L, Huang S, Du W, Zhu C, Xu H. Effects of the molecular weight and molar ratio of poly(2-ethyl-2-oxazoline)-based lipid on the pH sensitivity, stability, and antitumor efficacy of liposomes. Drug Dev Ind Pharm 2020; 46:283-295. [PMID: 31944130 DOI: 10.1080/03639045.2020.1717514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this study, we evaluated and screened the effects of the molecular weight (MW) and molar ratio of poly(2-ethyl-2-oxazoline)-cholesteryl methyl carbonate (PEtOz-CHMC) on the pH sensitivity, stability, and antitumor efficacy of liposomes. The pH sensitivity of PEtOz-CHMC with different MWs and molar ratios was screened by drug release and cytotoxicity experiments at different pH levels. Results indicated that the liposomes coated with PEtOz1k-CHMC (7% molar ratio) and PEtOz2k-CHMC (5% molar ratio) exhibited the desirable pH responsiveness. When the MW of PEtOz was relatively low, 7% of the modified ratio obtained the strongest stability, but the turbidity of the liposomes did not obviously change when the molar ratio of PEtOz-CHMC was further increased. A375 cells were used as models to investigate the cellular uptake and intracellular localization of coumarin-6-loaded liposomes (C6-L), PEGylated liposomes (PEG-C6-L), and PEtOzylated liposomes. PEtOz1k-C6-L and PEtOz2k-C6-L presented remarkably stronger fluorescence intensity at low pH than at pH 7.4, whereas C6-L and PEG-C6-L did not achieve any obvious diversity at different pH conditions. Compared with C6-L and PEG-C6-L, PEtOz-C6-L showed efficient intracellular trafficking, including endosomal/lysosomal escape and cytoplasmic release. Pharmacokinetic experiments demonstrated that half-lives of PEG2k-C6-L, PEtOz2k-C6-L, and PEtOz1k-C6-L were 11.89-, 7.00-, and 5.29-fold times higher than those of C6-L, respectively. Among the liposomes, the DOX·HCl-loaded liposomes coated with PEtOz2k-CHMC demonstrated the strongest antitumor efficacy against B16 tumor xenograft models in vivo. These findings provide the feasibility of using PEtOz-CHMC with optimal pH sensitivity and long circulation to extend the application of liposomes to efficient anticancer drug delivery.
Collapse
Affiliation(s)
- Luqiao Wen
- Department of Pharmacy, College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, People's Republic of China
| | - Shouzhen Huang
- Department of Pharmacy, College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, People's Republic of China
| | - Weiang Du
- Department of Pharmacy, College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, People's Republic of China
| | - Caili Zhu
- Department of Pharmacy, College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, People's Republic of China
| | - Huan Xu
- Department of Pharmacy, College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, People's Republic of China
| |
Collapse
|
22
|
Shukla RP, Dewangan J, Urandur S, Banala VT, Diwedi M, Sharma S, Agrawal S, Rath SK, Trivedi R, Mishra PR. Multifunctional hybrid nanoconstructs facilitate intracellular localization of doxorubicin and genistein to enhance apoptotic and anti-angiogenic efficacy in breast adenocarcinoma. Biomater Sci 2020; 8:1298-1315. [DOI: 10.1039/c9bm01246j] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The progressive development of tumors leading to angiogenesis marks the advancement of cancer which requires specific targeted treatment preferably with combination chemotherapy.
Collapse
Affiliation(s)
- Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Jayant Dewangan
- Division of Toxicology
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Sandeep Urandur
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Venkatesh Teja Banala
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Monika Diwedi
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Shweta Sharma
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Sristi Agrawal
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | | | - Ritu Trivedi
- Division of Endocrinology
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| |
Collapse
|
23
|
Negishi Y, Nomizu M. Laminin-derived peptides: Applications in drug delivery systems for targeting. Pharmacol Ther 2019; 202:91-97. [PMID: 31158392 DOI: 10.1016/j.pharmthera.2019.05.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/29/2022]
Abstract
Recently, the development of drug delivery systems (DDSs) for clinical application of anticancer drugs and gene therapy has rapidly progressed. In particular, DDS carriers used for chemotherapy and gene therapy are required to selectively deliver drugs and genes to cancer cells. Both the carrier and the molecule must in combination be highly selective in most cases. Possible candidate targeting molecules are the laminins, major basement membrane proteins that interact with various cells through their multiple constituent active peptide sequences. Laminin-derived peptides bind to various cellular receptors and have been used for DDSs as a targeting moiety. Here, we review the progress in laminin-derived peptide-conjugated DDSs. Drug and gene carriers as well as ultrasound diagnostic contrast agents utilizing laminin-derived peptides for selective targeting are useful components of DDSs and play important roles in cancer and in the neovasculature.
Collapse
Affiliation(s)
- Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan.
| |
Collapse
|
24
|
Shah VM, Nguyen DX, Patel P, Cote B, Al-Fatease A, Pham Y, Huynh MG, Woo Y, Alani AWG. Liposomes produced by microfluidics and extrusion: A comparison for scale-up purposes. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 18:146-156. [DOI: 10.1016/j.nano.2019.02.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/30/2019] [Accepted: 02/19/2019] [Indexed: 10/27/2022]
|
25
|
Mahendra A, James HP, Jadhav S. PEG-grafted phospholipids in vesicles: Effect of PEG chain length and concentration on mechanical properties. Chem Phys Lipids 2019; 218:47-56. [DOI: 10.1016/j.chemphyslip.2018.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/10/2018] [Accepted: 12/01/2018] [Indexed: 10/27/2022]
|
26
|
Chen W, Deng W, Xu X, Zhao X, Vo JN, Anwer AG, Williams TC, Cui H, Goldys EM. Photoresponsive endosomal escape enhances gene delivery using liposome-polycation-DNA (LPD) nanovectors. J Mater Chem B 2018; 6:5269-5281. [PMID: 32254764 DOI: 10.1039/c8tb00994e] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lipid-based nanocarriers with stimuli responsiveness have been utilized as controlled release systems for gene/drug delivery applications. In our work, by taking advantage of the high complexation capability of polycations and the light triggered properties, we designed a novel photoresponsive liposome-polycation-DNA (LPD) platform. This LPD carrier incorporates verteporfin (VP) in lipid bilayers and the complex of polyethylenimine (PEI)/plasmid DNA (pDNA) encoding EGFP (polyplex) in the central cavities of the liposomes. The liposomes were formulated with cationic lipids, PEGylated neutral lipids and cholesterol molecules, which improve their stability and cellular uptake in the serum-containing media. We evaluated the nanocomplex stability by monitoring size changes over six days, and the cellular uptake of the nanocomplex by imaging the intracellular route. We also demonstrated that light triggered the cytoplasmic release of pDNA upon irradiation with a 690 nm LED light source. Furthermore, this light triggered mechanism has been studied at the subcellular level. The activated release is driven by the generation of reactive oxygen species (ROS) from VP after light illumination. These ROS oxidize and destabilize the liposomal and endolysosomal membranes, leading to the release of pDNA into the cytosol and subsequent gene transfer activities. Light-triggered endolysosomal escape of pDNA at different time points was confirmed by a quantitative analysis of colocalization between pDNA and endolysosomes. The increased expression of the reporter EGFP in human colorectal cancer cells was also quantified after light illumination at various time points. The efficiency of this photo-induced gene transfection was demonstrated to be more than double compared to non-irradiated controls. Additionally, we observed a reduced cytotoxicity of the LPDs compared with the polyplexes alone. This study has thus shown that light-triggered and biocompatible LPDs enable an improved control of efficient gene delivery, which will be beneficial for future gene therapies.
Collapse
Affiliation(s)
- Wenjie Chen
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Department of Physics and Astronomy, Macquarie University, Sydney, New South Wales 2109, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Negishi Y, Hamano N, Sato H, Katagiri F, Takatori K, Endo-Takahashi Y, Kikkawa Y, Nomizu M. Development of a Screening System for Targeting Carriers Using Peptide-Modified Liposomes and Tissue Sections. Biol Pharm Bull 2018; 41:1107-1111. [DOI: 10.1248/bpb.b18-00151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Nobuhito Hamano
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Hinako Sato
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Fumihiko Katagiri
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Kyohei Takatori
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Yoko Endo-Takahashi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Yamato Kikkawa
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
28
|
PMO Delivery System Using Bubble Liposomes and Ultrasound Exposure for Duchenne Muscular Dystrophy Treatment. Methods Mol Biol 2018; 1687:185-192. [PMID: 29067664 DOI: 10.1007/978-1-4939-7374-3_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder characterized by progressive muscle degeneration, caused by nonsense or frameshift mutations in the dystrophin (DMD) gene. Antisense oligonucleotides can be used to induce specific exon skipping; recently, a phosphorodiamidate morpholino oligomer (PMO) has been approved for clinical use in DMD. However, an efficient PMO delivery strategy is required to improve the therapeutic efficacy in DMD patients. We previously developed polyethylene glycol (PEG)-modified liposomes containing ultrasound contrast gas, "Bubble liposomes" (BLs), and found that the combination of BLs with ultrasound exposure is a useful gene delivery tool. Here, we describe an efficient PMO delivery strategy using the combination of BLs and ultrasound exposure to treat muscles in a DMD mouse model (mdx). This ultrasound-mediated BL technique can increase the PMO-mediated exon-skipping efficiency, leading to significantly increased dystrophin expression. Thus, the combination of BLs and ultrasound exposure may be a feasible PMO delivery method to improve therapeutic efficacy and reduce the PMO dosage for DMD treatment.
Collapse
|
29
|
Chen J, Chen Y, Cheng Y, Gao Y. Glycyrrhetinic Acid Liposomes Containing Mannose-Diester Lauric Diacid-Cholesterol Conjugate Synthesized by Lipase-Catalytic Acylation for Liver-Specific Delivery. Molecules 2017; 22:molecules22101598. [PMID: 28946644 PMCID: PMC6151824 DOI: 10.3390/molecules22101598] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 09/07/2017] [Accepted: 09/17/2017] [Indexed: 11/29/2022] Open
Abstract
Mannose-diester lauric diacid-cholesterol (Man-DLD-Chol), as a liposomal target ligand, was synthesized by lipase catalyzed in a non-aqueous medium. Its chemical structure was confirmed by mass spectrometry (MS) and nuclear magnetic resonance (NMR) spectroscopy. Glycyrrhetinic acid (GA) liposomes containing Man-DLD-Chol (Man-DLD-Chol-GA-Lp) were prepared by the film-dispersion method. We evaluated the characterizations of liposomes, drug-release in vitro, the hemolytic test, cellular uptake, pharmacokinetics, and the tissue distributions. The cellular uptake in vitro suggested that the uptake of Man-DLD-Chol-modified liposomes was significantly higher than that of unmodified liposomes in HepG2 cells. Pharmacokinetic parameters indicated that Man-DLD-Chol-GA-Lp was eliminated more rapidly than GA-Lp. In tissue distributions, the targeting efficiency (Te) of Man-DLD-Chol-GA-Lp on liver was 54.67%, relative targeting efficiency (RTe) was 3.39, relative uptake rate (Re) was 4.78, and peak concentration ratio (Ce) was 3.46. All these results supported the hypothesis that Man-DLD-Chol would be an efficient liposomal carrier, and demonstrated that Man-DLD-Chol-GA-Lp has potential as a drug delivery for liver-targeting therapy.
Collapse
Affiliation(s)
- Jing Chen
- Shool of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Yuchao Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510115, China.
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, China.
- Postdoctoral Programme, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Yi Cheng
- Shool of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Youheng Gao
- Shool of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
30
|
Zhang M, Ma Y, Wang Z, Han Z, Gao W, Gu Y. Optimizing molecular weight of octyl chitosan as drug carrier for improving tumor therapeutic efficacy. Oncotarget 2017; 8:64237-64249. [PMID: 28969066 PMCID: PMC5609998 DOI: 10.18632/oncotarget.19452] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/26/2017] [Indexed: 02/01/2023] Open
Abstract
Macromolecular drug carriers have attracted much attention taking advantage of passive tumor targeting property and excellent biocompatibility. For many biomedical applications, however, the effectiveness of the carriers is insufficient, which complicate further development into clinical use. Here, we systematically investigated the effects of molecular weight (from 1KDa to 300KDa) of macromolecular drug carrier, octyl chitosan, on tumor accumulation and penetration, as well as drug loading and releasing profiles. It was found that the molecular weight of chitosan influenced the cellular uptake and pharmacokinetic behavior of the nanocarriers, which ultimately determined their drug delivery efficiency. Interestingly, increased molecular weight of chitosan decreased its cellular uptake but increased its resident time in blood, which provided ample time for tumor accumulation. Moreover, the molecular weight altered the drug loading capability and release profile. Our results demonstrated that 10KDa octyl chitosan was an ideal candidate for anticancer drug delivery, which could deliver anticancer agent to tumor tissues and release drugs in tumor cells more effectively than those of other molecular weights, and finally result in better therapeutic effect.
Collapse
Affiliation(s)
- Min Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Ma
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhaohui Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhihao Han
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Weidong Gao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yueqing Gu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
31
|
Zeng F, Ju RJ, Liu L, Xie HJ, Mu LM, Zhao Y, Yan Y, Hu YJ, Wu JS, Lu WL. Application of functional vincristine plus dasatinib liposomes to deletion of vasculogenic mimicry channels in triple-negative breast cancer. Oncotarget 2017; 6:36625-42. [PMID: 26429872 PMCID: PMC4742200 DOI: 10.18632/oncotarget.5382] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 09/16/2015] [Indexed: 01/03/2023] Open
Abstract
Standard chemotherapy cannot eradicate triple-negative breast cancer (TNBC) while the residual cancer cells readily form the vasculogenic mimicry (VM) channels, which lead to the relapse of cancer after treatment. In this study, the functional vincristine plus dasatinib liposomes, modified by a targeting molecule DSPE-PEG2000-c(RGDyK), were fabricated to address this issue. The investigations were performed on TNBC MDA-MB-231 cells and MDA-MB-231 xenografts in nude mice. The liposomes exhibited the superior performances in the following aspects: the enhancement of cellular uptake via targeted action; the induction of apoptosis via activation of caspase 8, 9, and 3, increased expression of Bax, decreased expression of Mcl-1, and generation of reactive oxygen species (ROS); and the deletion of VM channels via inhibitions on the VM channel indicators, which consisted of vascular endothelial-cadherin (VE-Cad), focal adhesion kinase (FAK), phosphatidylinositide 3-kinase (PI3K), and matrix metallopeptidases (MMP-2, and MMP-9). Furthermore, the liposomes displayed the prolonged circulation time in the blood, the increased accumulation in tumor tissue, and the improved therapeutic efficacy along with deletion of VM channels in the TNBC-bearing mice. In conclusion, the nanostructured functional drug-loaded liposomes may provide a promising strategy for the treatment of invasive TNBC along with deletion of VM channels.
Collapse
Affiliation(s)
- Fan Zeng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Rui-Jun Ju
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lei Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hong-Jun Xie
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Li-Min Mu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yao Zhao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yan Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ying-Jie Hu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jia-Shuan Wu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wan-Liang Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
32
|
Elsaid Z, Taylor KMG, Puri S, Eberlein CA, Al-Jamal K, Bai J, Klippstein R, Wang JTW, Forbes B, Chana J, Somavarapu S. Mixed micelles of lipoic acid-chitosan-poly(ethylene glycol) and distearoylphosphatidylethanolamine-poly(ethylene glycol) for tumor delivery. Eur J Pharm Sci 2017; 101:228-242. [PMID: 28163163 DOI: 10.1016/j.ejps.2017.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/27/2017] [Accepted: 02/01/2017] [Indexed: 01/21/2023]
Abstract
Many chemotherapeutics suffer from poor aqueous solubility and tissue selectivity. Distearoylphosphatidylethanolamine-poly(ethylene glycol) (DSPE-PEG) micelles are a promising formulation strategy for the delivery of hydrophobic anticancer drugs. However, storage and in vivo instability restrict their use. The aim of this study was to prepare mixed micelles, containing a novel polymer, lipoic acid-chitosan-poly(ethylene glycol) (LACPEG), and DSPE-PEG, to overcome these limitations and potentially increase cancer cell internalisation. Drug-loaded micelles were prepared with a model tyrosine kinase inhibitor and characterized for size, surface charge, stability, morphology, drug entrapment efficiency, cell viability (A549 and PC-9 cell lines), in vivo biodistribution, ex vivo tumor accumulation and cellular internalisation. Micelles of size 30-130nm with entrapment efficiencies of 46-81% were prepared. LACPEG/DSPE-PEG mixed micelles showed greater interaction with the drug (condensing to half their size following entrapment), greater stability, and a safer profile in vitro compared to DSPE-PEG micelles. LACPEG/DSPE-PEG and DSPE-PEG micelles had similar entrapment efficiencies and in vivo tumor accumulation levels, but LACPEG/DSPE-PEG micelles showed higher tumor cell internalisation. Collectively, these findings suggest that LACPEG/DSPE-PEG mixed micelles provide a promising platform for tumor delivery of hydrophobic drugs.
Collapse
Affiliation(s)
- Zeeneh Elsaid
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom.
| | - Kevin M G Taylor
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Sanyogitta Puri
- AstraZeneca, Macclesfield, Cheshire East SK10 2NA, United Kingdom
| | - Cath A Eberlein
- AstraZeneca, Macclesfield, Cheshire East SK10 2NA, United Kingdom
| | - Khuloud Al-Jamal
- Kings College London, Franklin-Wilkins Building, Stamford Street, London SE1 8WA, United Kingdom
| | - Jie Bai
- Kings College London, Franklin-Wilkins Building, Stamford Street, London SE1 8WA, United Kingdom
| | - Rebecca Klippstein
- Kings College London, Franklin-Wilkins Building, Stamford Street, London SE1 8WA, United Kingdom
| | - Julie Tzu-Wen Wang
- Kings College London, Franklin-Wilkins Building, Stamford Street, London SE1 8WA, United Kingdom
| | - Ben Forbes
- Kings College London, Franklin-Wilkins Building, Stamford Street, London SE1 8WA, United Kingdom
| | - Jasminder Chana
- Kings College London, Franklin-Wilkins Building, Stamford Street, London SE1 8WA, United Kingdom
| | | |
Collapse
|
33
|
Tang HX, Zhao TW, Zheng T, Sheng YJ, Zheng HS, Zhang YS. Liver-targeting liposome drug delivery system and its research progress in liver diseases. Shijie Huaren Xiaohua Zazhi 2016; 24:4238-4246. [DOI: 10.11569/wcjd.v24.i31.4238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Liposome-based targeted therapy is mainly divided into active targeting, passive targeting, and physical and chemical targeting. In terms of liver targeting, because of specificity, active liver-targeting liposomes have received more and more attention, and these types of liposomes can be used in liver fibrosis, hepatitis and other chronic liver diseases. In addition, the particle size could control the passive liver targeting of liposomes, while the liver-targeted liposomes of the physical and chemical targeting type have advantages in treating hepatic carcinoma. In this paper, we focus on the basics and application of liver-targeting liposome drug delivery system in hepatic diseases.
Collapse
|
34
|
Zhang Y, Mintzer E, Uhrich KE. Synthesis and characterization of PEGylated bolaamphiphiles with enhanced retention in liposomes. J Colloid Interface Sci 2016; 482:19-26. [DOI: 10.1016/j.jcis.2016.07.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 10/21/2022]
|
35
|
Zhang J, Chen Y, Li X, Liang X, Luo X. The influence of different long-circulating materials on the pharmacokinetics of liposomal vincristine sulfate. Int J Nanomedicine 2016; 11:4187-97. [PMID: 27616886 PMCID: PMC5008646 DOI: 10.2147/ijn.s109547] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PURPOSE This study was designed to improve the in vivo pharmacokinetics of long-circulating vincristine sulfate (VS)-loaded liposomes; three different long-circulating materials, chitosan, poly(ethylene glycol)-1,2-distearoyl sn-glycero-3-phosphatidylethanolamine (PEG-DSPE), and poly(ethylene glycol)-poly-lactide-co-glycolide (PEG-PLGA), were evaluated at the same coating molar ratio with the commercial product Marqibo(®) (vincristine sulfate liposome injection [VSLI]). MATERIALS AND METHODS VS-loaded liposomes were prepared by a pH gradient method and were then coated with chitosan, PEG-DSPE, or PEG-PLGA. Physicochemical properties, including the morphology, particle size, zeta potential, encapsulation efficiency (EE%), pH, drug loading, and in vitro release, were determined. Preservation stability and pharmacokinetic studies were performed to compare the membrane-coated liposomes with either commercially available liposomes or the VS solution. RESULTS The sphere-like morphology of the vesicles was confirmed by transmission electron microscope. Increased particle size, especially for the chitosan formulation, was observed after the coating process. However, the EE% was ~99.0% with drug loading at 2.0 mg/mL, which did not change after the coating process. The coating of long-circulation materials, except for chitosan, resulted in negatively charged and stable vesicles at physiological pH. The near-zero zeta potential exhibited by the PEG-DSPE formulation leads to a longer circulation lifetime and improved absorption for VS, when compared with the PEG-PLGA formulation. Compared with the commercial product, PEG was responsible for a higher plasma VS concentration and a longer half-life. CONCLUSION PEG-DSPE coating may be related to better absorption, based on the stability and a pharmacokinetic improvement in the blood circulation time.
Collapse
Affiliation(s)
- Jing Zhang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education
| | - Yingchong Chen
- Key Laboratory of Modern Preparation of TCM, Ministry of Education
| | - Xiang Li
- Key Laboratory of Modern Preparation of TCM, Ministry of Education; State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, People's Republic of China
| | - Xinli Liang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education
| | - Xiaojian Luo
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, People's Republic of China
| |
Collapse
|
36
|
Lee H, Larson RG. Adsorption of Plasma Proteins onto PEGylated Lipid Bilayers: The Effect of PEG Size and Grafting Density. Biomacromolecules 2016; 17:1757-65. [DOI: 10.1021/acs.biomac.6b00146] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Hwankyu Lee
- Department
of Chemical Engineering, Dankook University, Yongin, 448-701, South Korea
| | - Ronald G. Larson
- Department
of Chemical Engineering, Biomedical Engineering, Mechanical Engineering,
and Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
37
|
Negishi Y, Tsunoda Y, Hamano N, Omata D, Endo-Takahashi Y, Suzuki R, Maruyama K, Nomizu M, Aramaki Y. Ultrasound-mediated gene delivery systems by AG73-modified Bubble liposomes. Biopolymers 2016; 100:402-7. [PMID: 23532952 DOI: 10.1002/bip.22246] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/11/2013] [Accepted: 03/17/2013] [Indexed: 11/06/2022]
Abstract
Targeted gene delivery to neovascular vessels in tumors is considered a promising strategy for cancer therapy. We previously reported that "Bubble liposomes" (BLs), which are ultrasound (US) imaging gas-encapsulating liposomes, were suitable for US imaging and gene delivery. When BLs are exposed to US, the bubble is destroyed, creating a jet stream by cavitation, and resulting in the instantaneous ejection of extracellular plasmid DNA (pDNA) or other nucleic acids into the cytosol. We developed AG73 peptide-modified Bubble liposomes (AG73-BL) as a targeted US contrast agent, which was designed to attach to neovascular tumor vessels and to allow specific US detection of angiogenesis (Negishi et al., Biomaterials 2013, 34, 501-507). In this study, to evaluate the effectiveness of AG73-BL as a gene delivery tool for neovascular vessels, we examined the gene transfection efficiency of AG73-BL with US exposure in primary human endothelial cells (HUVEC). The transfection efficiency was significantly enhanced if the AG73-BL attached to the HUVEC was exposed to US compared to the BL-modified with no peptide or scrambled peptide. In addition, the cell viability was greater than 80% after transfection with AG73-BL. These results suggested that after the destruction of the AG73-BL with US exposure, a cavitation could be effectively induced by the US exposure against AG73-BL binding to the cell surface of the HUVEC, and the subsequent gene delivery into cells could be enhanced. Thus, AG73-BL may be useful for gene delivery as well as for US imaging of neovascular vessels.
Collapse
Affiliation(s)
- Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Han NK, Shin DH, Kim JS, Weon KY, Jang CY, Kim JS. Hyaluronan-conjugated liposomes encapsulating gemcitabine for breast cancer stem cells. Int J Nanomedicine 2016; 11:1413-25. [PMID: 27103799 PMCID: PMC4827594 DOI: 10.2147/ijn.s95850] [Citation(s) in RCA: 39] [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] [Indexed: 12/15/2022] Open
Abstract
Investigation of potential therapeutics for targeting breast cancer stem cells (BCSCs) is important because these cells are regarded as culprit of breast cancer relapse. Accomplishing this kind of strategy requires a specific drug-delivery system using the distinct features of liposomes. Studies on targeted liposomal delivery systems have indicated the conjugation of hyaluronan (HA), a primary ligand for CD44 surface markers, as an appropriate method for targeting BCSCs. For this study, enriched BCSCs were obtained by culturing MCF-7 breast cancer cells in nonadherent conditions. The enriched BCSCs were challenged with HA-conjugated liposomes encapsulating gemcitabine (2, 2-difluoro-2-deoxycytidine, GEM). In vitro study showed that the HA-conjugated liposomes significantly enhanced the cytotoxicity, anti-migration, and anti-colony formation abilities of GEM through targeting of CD44 expressed on BCSCs. In pharmacokinetic study, area under the drug concentration vs time curve (AUC) of the immunoliposomal GEM was 3.5 times higher than that of free GEM, indicating that the HA-conjugated liposomes enhanced the stability of GEM in the bloodstream and therefore prolonged its half-life time. The antitumor effect of the immunoliposomal GEM was 3.3 times higher than that of free GEM in a xenograft mouse model, probably reflecting the unique targeting of the CD44 receptor by HA and the increased cytotoxicity and stability through the liposomal formulation. Furthermore, marginal change in body weight demonstrated that the use of liposomes considerably reduced the systemic toxicity of GEM on normal healthy cells. Taken together, this study demonstrates that HA-conjugated liposomes encapsulating GEM show promise for the therapy of breast cancer in vitro and in a xenograft model by targeting the BCSCs.
Collapse
Affiliation(s)
- Na-Kyung Han
- Research Center for Cell Fate Control (RCCFC) and College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| | - Dae Hwan Shin
- Research Center for Cell Fate Control (RCCFC) and College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| | - Jung Seok Kim
- Research Center for Cell Fate Control (RCCFC) and College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| | - Kwon Yeon Weon
- College of Pharmacy, Catholic University of Daegu, Gyeongbuk, Korea
| | - Chang-Young Jang
- Research Center for Cell Fate Control (RCCFC) and College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| | - Jin-Seok Kim
- Research Center for Cell Fate Control (RCCFC) and College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
| |
Collapse
|
39
|
Enhanced anti-tumor efficacy and safety profile of tumor microenvironment-responsive oncolytic adenovirus nanocomplex by systemic administration. Acta Biomater 2015; 28:86-98. [PMID: 26365317 DOI: 10.1016/j.actbio.2015.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 08/29/2015] [Accepted: 09/09/2015] [Indexed: 02/04/2023]
Abstract
Oncolytic adenovirus (Ad) holds great promise as a potential gene therapy for cancer. However, intravenously administered Ad may encounter difficulties due to unfavorable host responses, non-specific interactions, and the heterogeneity of the tumor cell population. As an approach to combine the advantages of oncolytic Ad and synthetic polymers and to address the associated difficulties, Ad was physically complexed with a pH-sensitive block copolymer, methoxy poly(ethylene glycol)-b-poly(l-histidine) (mPEG-b-pHis). The in vitro transduction efficiency at an acidic extracellular pH was remarkably enhanced in cancer cells when treated with the Ad expressing green fluorescent protein (GFP) coated with mPEG-b-pHis (c-dE1/GFP) as compared to that of naked Ad (n-dE1/GFP). Time-lapse total internal reflection fluorescence microscopic imaging revealed a significantly enhanced cellular uptake rate of c-dE1/GFP at acidic tumor pH when compared with that at neutral pH or naked cognate Ad (n-dE1/GFP). In addition, c-dE1/GFP remained relatively stable in human serum-containing media, and considerably reduced both the innate and adaptive immune response against Ad. Moreover, the therapeutic efficacy and survival benefit of mPEG-b-pHis-complexed oncolytic Ad (c-H5mT/Luc) by systemic treatment was significantly enhanced compared to that with naked oncolytic Ad (n-H5mT/Luc) in both coxsackie and adenovirus receptor-positive and -negative tumors. Whole-body bioluminescence imaging showed 7.3-fold higher luciferase expression at the tumor site and 23.0-fold less luciferase expression in liver tissue for c-H5mT/Luc relative to that for naked oncolytic Ad (n-H5mT/Luc). Considering the heterogeneity of tumor tissue, these results are important for guiding the development of more potent and specific treatment of devastating metastatic cancers using this viral system. STATEMENT OF SIGNIFICANCE Although adenoviral systems have shown considerable promise and undergone extensive evaluation attempts to specifically target Ad vectors to cancer cells have met limited success. This shortcoming is due to the strong immune response stimulated by Ad and the hepatotoxicity of the viral particles. To overcome restricted vector issues, we generated Ad/mPEG-b-pHis for tumor microenvironment-targeting hybrid vector systems, an oncolytic Ad coated with a pH-responsive polymer, mPEG-b-pHis. The Ad/mPEG-b-pHis exhibited pH-dependent transduction efficiency and cancer-cell killing effects. Moreover, systemic administration of oncolytic Ad/mPEG-b-pHis led to marked suppression of tumor growth and tumor-specific viral replication. Ad successfully avoided the innate and adaptive immune responses and liver accumulation with the help of mPEG-b-pHis on its surface.
Collapse
|
40
|
Enhancement of Blood-Brain Barrier Permeability and Delivery of Antisense Oligonucleotides or Plasmid DNA to the Brain by the Combination of Bubble Liposomes and High-Intensity Focused Ultrasound. Pharmaceutics 2015; 7:344-62. [PMID: 26402694 PMCID: PMC4588205 DOI: 10.3390/pharmaceutics7030344] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/03/2015] [Accepted: 09/14/2015] [Indexed: 12/29/2022] Open
Abstract
The blood–brain barrier (BBB) is a major obstacle that prevents therapeutic drugs or genes from being delivered to the central nervous system. Therefore, it is important to develop methods to enhance the permeability of the BBB. We have developed echo-contrast gas (C3F8) entrapping liposomes (Bubble liposomes, BLs) that can work as a gene delivery tool in combination with ultrasound (US) exposure. Here, we studied whether the permeability of the BBB can be enhanced by the combination of BLs and high-intensity focused ultrasound (HIFU). Mice were intravenously injected with Evans blue (EB). BLs were subsequently injected, and the right hemispheres were exposed to HIFU. As a result, the accumulation of EB in the HIFU-exposed brain hemispheres was increased over that observed in the non-HIFU-exposed hemispheres, depending on the intensity and the duration of the HIFU. Similarly, the combination of BLs and HIFU allowed fluorescent-labeled antisense oligonucleotides to be delivered into the HIFU-exposed left hemispheres of the treated mice. Furthermore, a firefly luciferase-expressing plasmid DNA was delivered to the brain by the combination method of BLs and HIFU, which resulted in the increased gene expression in the brain at the focused-US exposure site. These results suggest that the method of combining BLs and HIFU together serves as a useful means for accelerating the permeability of BBB and thereby enabling antisense oligonucleotides or genes to be delivered to the focused brain site.
Collapse
|
41
|
Tian C, Ling J, Shen YQ. Self-assembly and pH-responsive properties of poly(L-glutamic acid-r-L-leucine) and poly(L-glutamic acid-r-L-leucine)-b-polysarcosine. CHINESE JOURNAL OF POLYMER SCIENCE 2015. [DOI: 10.1007/s10118-015-1669-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
42
|
Lee KL, Shukla S, Wu M, Ayat NR, El Sanadi CE, Wen AM, Edelbrock JF, Pokorski JK, Commandeur U, Dubyak GR, Steinmetz NF. Stealth filaments: Polymer chain length and conformation affect the in vivo fate of PEGylated potato virus X. Acta Biomater 2015; 19:166-79. [PMID: 25769228 PMCID: PMC4411193 DOI: 10.1016/j.actbio.2015.03.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 02/22/2015] [Accepted: 03/03/2015] [Indexed: 12/23/2022]
Abstract
Nanoparticles hold great promise for delivering medical cargos to cancerous tissues to enhance contrast and sensitivity of imaging agents or to increase specificity and efficacy of therapeutics. A growing body of data suggests that nanoparticle shape, in combination with surface chemistry, affects their in vivo fates, with elongated filaments showing enhanced tumor targeting and tissue penetration, while promoting immune evasion. The synthesis of high aspect ratio filamentous materials at the nanoscale remains challenging using synthetic routes; therefore we turned toward nature's materials, developing and studying the filamentous structures formed by the plant virus potato virus X (PVX). We recently demonstrated that PVX shows enhanced tumor homing in various preclinical models. Like other nanoparticle systems, the proteinaceous platform is cleared from circulation and tissues by the mononuclear phagocyte system (MPS). To increase bioavailability we set out to develop PEGylated stealth filaments and evaluate the effects of PEG chain length and conformation on pharmacokinetics, biodistribution, as well as potential immune and inflammatory responses. We demonstrate that PEGylation effectively reduces immune recognition while increasing pharmacokinetic profiles. Stealth filaments show reduced interaction with cells of the MPS; the protein:polymer hybrids are cleared from the body tissues within hours to days indicating biodegradability and biocompatibility. Tissue compatibility is indicated with no apparent inflammatory signaling in vivo. Tailoring PEG chain length and conformation (brush vs. mushroom) allows tuning of the pharmacokinetics, yielding long-circulating stealth filaments for applications in nanomedicine.
Collapse
Affiliation(s)
- Karin L Lee
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States
| | - Sourabh Shukla
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States
| | - Mengzhi Wu
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States
| | - Nadia R Ayat
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States
| | - Caroline E El Sanadi
- Department of Physiology and Biophysics, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States
| | - Amy M Wen
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States
| | - John F Edelbrock
- Department of Macromolecular Science and Engineering, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States
| | - Jonathan K Pokorski
- Department of Macromolecular Science and Engineering, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States
| | - Ulrich Commandeur
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringer Weg 1, 52074 Aachen, Germany
| | - George R Dubyak
- Department of Physiology and Biophysics, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States
| | - Nicole F Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States; Department of Macromolecular Science and Engineering, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States; Department of Radiology, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States; Department of Materials Science and Engineering, Case Western Reserve University Schools of Medicine and Engineering, Cleveland, OH 44106, United States.
| |
Collapse
|
43
|
Affiliation(s)
- Bhushan S Pattni
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States
| | - Vladimir V Chupin
- Laboratory for Advanced Studies of Membrane Proteins, Moscow Institute of Physics and Technology , Dolgoprudny 141700, Russia
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States.,Department of Biochemistry, Faculty of Science, King Abdulaziz University , Jeddah 21589, Saudi Arabia
| |
Collapse
|
44
|
Li K, Liu B. Polymer-encapsulated organic nanoparticles for fluorescence and photoacoustic imaging. Chem Soc Rev 2015; 43:6570-97. [PMID: 24792930 DOI: 10.1039/c4cs00014e] [Citation(s) in RCA: 681] [Impact Index Per Article: 68.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Polymer encapsulated organic nanoparticles have recently attracted increasing attention in the biomedical field because of their unique optical properties, easy fabrication and outstanding performance as imaging and therapeutic agents. Of particular importance is the polymer encapsulated nanoparticles containing conjugated polymers (CP) or fluorogens with aggregation induced emission (AIE) characteristics as the core, which have shown significant advantages in terms of tunable brightness, superb photo- and physical stability, good biocompatibility, potential biodegradability and facile surface functionalization. In this review, we summarize the latest advances in the development of polymer encapsulated CP and AIE fluorogen nanoparticles, including preparation methods, material design and matrix selection, nanoparticle fabrication and surface functionalization for fluorescence and photoacoustic imaging. We also discuss their specific applications in cell labeling, targeted in vitro and in vivo imaging, blood vessel imaging, cell tracing, inflammation monitoring and molecular imaging. We specially focus on strategies to fine-tune the nanoparticle property (e.g. size and fluorescence quantum yield) through precise engineering of the organic cores and careful selection of polymer matrices. The review also highlights the merits and limitations of these nanoparticles as well as strategies used to overcome the limitations. The challenges and perspectives for the future development of polymer encapsulated organic nanoparticles are also discussed.
Collapse
Affiliation(s)
- Kai Li
- Institute of Materials Research and Engineering, A*STAR, 3 Research Link, Singapore 117602.
| | | |
Collapse
|
45
|
Lazarovits J, Chen YY, Sykes EA, Chan WCW. Nanoparticle–blood interactions: the implications on solid tumour targeting. Chem Commun (Camb) 2015; 51:2756-67. [DOI: 10.1039/c4cc07644c] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This review examines nanoparticle–blood interactions, their implications on solid tumour targeting, and provides an outlook to guide future nanoparticle design.
Collapse
Affiliation(s)
- James Lazarovits
- Institute of Biomaterials and Biomedical Engineering
- University of Toronto
- Toronto
- Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research
| | - Yih Yang Chen
- Institute of Biomaterials and Biomedical Engineering
- University of Toronto
- Toronto
- Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research
| | - Edward A. Sykes
- Institute of Biomaterials and Biomedical Engineering
- University of Toronto
- Toronto
- Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research
| | - Warren C. W. Chan
- Institute of Biomaterials and Biomedical Engineering
- University of Toronto
- Toronto
- Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research
| |
Collapse
|
46
|
Glycosylation-mediated targeting of carriers. J Control Release 2014; 190:542-55. [DOI: 10.1016/j.jconrel.2014.06.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 05/29/2014] [Accepted: 06/02/2014] [Indexed: 12/24/2022]
|
47
|
Tomuleasa C, Braicu C, Irimie A, Craciun L, Berindan-Neagoe I. Nanopharmacology in translational hematology and oncology. Int J Nanomedicine 2014; 9:3465-79. [PMID: 25092977 PMCID: PMC4113407 DOI: 10.2147/ijn.s60488] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nanoparticles have displayed considerable promise for safely delivering therapeutic agents with miscellaneous therapeutic properties. Current progress in nanotechnology has put forward, in the last few years, several therapeutic strategies that could be integrated into clinical use by using constructs for molecular diagnosis, disease detection, cytostatic drug delivery, and nanoscale immunotherapy. In the hope of bringing the concept of nanopharmacology toward a viable and feasible clinical reality in a cancer center, the present report attempts to present the grounds for the use of cell-free nanoscale structures for molecular therapy in experimental hematology and oncology.
Collapse
Affiliation(s)
- Ciprian Tomuleasa
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania ; Department of Hematology, Ion Chiricuta Cancer Center, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandra Irimie
- Department of Prosthetic Dentistry and Dental Materials, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucian Craciun
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania ; Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania ; Department of Functional Genomics and Experimental Pathology, the Oncological Institute "Prof Dr Ion Chiricuta", Cluj-Napoca, Romania
| |
Collapse
|
48
|
Novel resveratrol and 5-fluorouracil coencapsulated in PEGylated nanoliposomes improve chemotherapeutic efficacy of combination against head and neck squamous cell carcinoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:424239. [PMID: 25114900 PMCID: PMC4119704 DOI: 10.1155/2014/424239] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/27/2014] [Indexed: 12/23/2022]
Abstract
Increasing consumption of tobacco and alcohol has led to a steady increase in the incidence of head and neck cancers in Asia. The drawbacks associated with the existing chemotherapeutic and surgical interventions have necessitated the development of a safer alternative for therapy of head and neck cancers. In this study we have explored the synergistic therapeutic potential of a phytochemical and chemotherapeutic agent using PEGylated liposomes as a delivery vehicle. Resveratrol and 5-fluorouracil were successfully coencapsulated in a single PEGylated nanoliposome. The thermal analysis and the nuclear magnetic resonance results revealed that resveratrol localized near the glycerol backbone of the liposomal membrane while 5-fluorouracil localized closer to the phosphate moiety, which influenced the release kinetics of both drugs. The nanoformulation was tested in vitro on a head and neck cancer cell line NT8e and was found to exhibit a GI50 similar to that of free 5-fluorouracil. Further, gene expression studies showed that the combination of resveratrol and 5-fluorouracil exhibited different effects on different genes that may influence the net antagonistic effect. The coencapsulation of resveratrol and 5-fluorouracil in a liposomal nanocarrier improved the cytotoxicity in comparison with the free drug combination when tested in vitro.
Collapse
|
49
|
Rüger R, Tansi FL, Rabenhold M, Steiniger F, Kontermann RE, Fahr A, Hilger I. In vivo near-infrared fluorescence imaging of FAP-expressing tumors with activatable FAP-targeted, single-chain Fv-immunoliposomes. J Control Release 2014; 186:1-10. [PMID: 24810115 DOI: 10.1016/j.jconrel.2014.04.050] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/17/2014] [Accepted: 04/24/2014] [Indexed: 12/01/2022]
Abstract
Molecular and cellular changes that precede the invasive growth of solid tumors include the release of proteolytic enzymes and peptides in the tumor stroma, the recruitment of phagocytic and lymphoid infiltrates and alteration of the extracellular matrix. The reactive tumor stroma consists of a large number of myofibroblasts, characterized by high expression of fibroblast activation protein alpha (FAP). FAP, a type-II transmembrane sialoglycoprotein is an attractive target in diagnosis and therapy of several pathologic disorders especially cancer. In the underlying work, a fluorescence-activatable liposome (fluorescence-quenched during circulation and fluorescence activation upon cellular uptake), bearing specific single-chain Fv fragments directed against FAP (scFv'FAP) was developed, and its potential for use in fluorescence diagnostic imaging of FAP-expressing tumor cells was evaluated by whole body fluorescence imaging. The liposomes termed anti-FAP-IL were prepared via post-insertion of ligand-phospholipid-conjugates into preformed DY-676-COOH-containing liposomes. The anti-FAP-IL revealed a homogeneous size distribution and showed specific interaction and binding with FAP-expressing cells in vitro. The high level of fluorescence quenching of the near-infrared fluorescent dye sequestered in the aqueous interior of the liposomes enables fluorescence imaging exclusively upon uptake and degradation by cells, which results in fluorescence activation. Only FAP-expressing cells were able to take up and activate fluorescence of anti-FAP-IL in vitro. Furthermore, anti-FAP-IL accumulated selectively in FAP-expressing xenograft models in vivo, as demonstrated by blocking experiments using free scFv'FAP. The local tumor fluorescence intensities were in agreement with the intrinsic degree of FAP-expression in different xenograft models. Thus, anti-FAP-IL can serve as a suitable in vivo diagnostic tool for pathological disorders accompanied by high FAP-expression.
Collapse
Affiliation(s)
- Ronny Rüger
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany.
| | - Felista L Tansi
- Dept. of Experimental Radiology, Institute of Diagnostic and Interventional Radiology I, Jena University Hospital-Friedrich Schiller University Jena, Erlanger Allee 101, 07747 Jena, Germany.
| | - Markus Rabenhold
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany
| | - Frank Steiniger
- Center for Electron Microscopy, Jena University Hospital-Friedrich Schiller University Jena, Ziegelmuehlenweg 1, 07743 Jena, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Alfred Fahr
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany.
| | - Ingrid Hilger
- Dept. of Experimental Radiology, Institute of Diagnostic and Interventional Radiology I, Jena University Hospital-Friedrich Schiller University Jena, Erlanger Allee 101, 07747 Jena, Germany.
| |
Collapse
|
50
|
Yue X, Dai Z. Recent advances in liposomal nanohybrid cerasomes as promising drug nanocarriers. Adv Colloid Interface Sci 2014; 207:32-42. [PMID: 24368133 DOI: 10.1016/j.cis.2013.11.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 11/04/2013] [Accepted: 11/18/2013] [Indexed: 10/25/2022]
Abstract
Liposomes have been extensively investigated as possible carriers for diagnostic or therapeutic agents due to their unique properties. However, liposomes still have not attained their full potential as drug and gene delivery vehicles because of their insufficient morphological stability. Recently, a super-stable and freestanding hybrid liposomal cerasome (partially ceramic- or silica-coated liposome) has drawn much attention as a novel drug delivery system because its atomic layer of polyorganosiloxane surface imparts higher morphological stability than conventional liposomes and its liposomal bilayer structure reduces the overall rigidity and density greatly compared to silica nanoparticles. Cerasomes are more biocompatible than silica nanoparticles due to the incorporation of the liposomal architecture into cerasomes. Cerasomes combine the advantages of both liposomes and silica nanoparticles but overcome their disadvantages so cerasomes are ideal drug delivery systems. The present review will first highlights some of the key advances of the past decade in the technology of cerasome production and then review current biomedical applications of cerasomes, with a view to stimulating further research in this area of study.
Collapse
|