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
|
Hadi Barhaghtalab R, Tanimowo Aiyelabegan H, Maleki H, Mirzavi F, Gholizadeh Navashenaq J, Abdi F, Ghaffari F, Vakili-Ghartavol R. Recent advances with erythrocytes as therapeutics carriers. Int J Pharm 2024; 665:124658. [PMID: 39236775 DOI: 10.1016/j.ijpharm.2024.124658] [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: 06/06/2024] [Revised: 08/24/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024]
Abstract
Erythrocytes have gained popularity as a natural option for in vivo drug delivery due to their advantages, which include lengthy circulation times, biocompatibility, and biodegradability. Consequently, the drug's pharmacokinetics and pharmacodynamics in red blood cells can be considerably up the dosage. Here, we provide an overview of the erythrocyte membrane's structure and discuss the characteristics of erythrocytes that influence their suitability as carrier systems. We also cover current developments in the erythrocyte-based nanocarrier, which could be used for both active and passive targeting of disease tissues, particularly those of the reticuloendothelial system (RES) and cancer tissues. We also go over the most recent discoveries about the in vivo and in vitro uses of erythrocytes for medicinal and diagnostic purposes. Moreover, the clinical relevance of erythrocytes is discussed in order to improve comprehension and enable the potential use of erythrocyte carriers in the management of various disorders.
Collapse
Affiliation(s)
| | | | - Hassan Maleki
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Fereshteh Abdi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Faezeh Ghaffari
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roghayyeh Vakili-Ghartavol
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Imperlini E, Di Marzio L, Cevenini A, Costanzo M, Nicola d'Avanzo, Fresta M, Orrù S, Celia C, Salvatore F. Unraveling the impact of different liposomal formulations on the plasma protein corona composition might give hints on the targeting capability of nanoparticles. NANOSCALE ADVANCES 2024; 6:4434-4449. [PMID: 39170967 PMCID: PMC11334990 DOI: 10.1039/d4na00345d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/27/2024] [Indexed: 08/23/2024]
Abstract
Nanoparticles (NPs) interact with biological fluids after being injected into the bloodstream. The interactions between NPs and plasma proteins at the nano-bio interface affect their biopharmaceutical properties and distribution in the organ and tissues due to protein corona (PrC) composition, and in turn, modification of the resulting targeting capability. Moreover, lipid and polymer NPs, at their interface, affect the composition of PrC and the relative adsorption and abundance of specific proteins. To investigate this latter aspect, we synthesized and characterized different liposomal formulations (LFs) with lipids and polymer-conjugated lipids at different molar ratios, having different sizes, size distributions and surface charges. The PrC composition of various designed LFs was evaluated ex vivo in human plasma by label-free quantitative proteomics. We also correlated the relative abundance of identified specific proteins in the coronas of the different LFs with their physicochemical properties (size, PDI, zeta potential). The evaluation of outputs from different bioinformatic tools discovered protein clusters allowing to highlight: (i) common as well as the unique species for the various formulations; (ii) correlation between each identified PrC and the physicochemical properties of LFs; (iii) some preferential binding determined by physicochemical properties of LFs; (iv) occurrence of formulation-specific protein patterns in PrC. Investigating specific clusters in PrC will help decode the multivalent roles of the protein pattern components in the drug delivery process, taking advantage of the bio-nanoscale recognition and identification for significant advances in nanomedicine.
Collapse
Affiliation(s)
- Esther Imperlini
- Department for Innovation in Biological, Agrofood and Forest Systems, University of Tuscia Viterbo 01100 Italy
| | - Luisa Di Marzio
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio" Via dei Vestini 31 66100 Chieti Italy +39 0871 3554711
| | - Armando Cevenini
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II Naples 80131 Italy +39 3356069177
- CEINGE-Biotecnologie Avanzate Franco Salvatore Naples 80145 Italy +39 081 3737880
| | - Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II Naples 80131 Italy +39 3356069177
- CEINGE-Biotecnologie Avanzate Franco Salvatore Naples 80145 Italy +39 081 3737880
| | - Nicola d'Avanzo
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia" Viale "S. Venuta" 88100 Catanzaro Italy
- Department of Experimental and Clinical Medicine, Research Center "ProHealth Translational Hub", "Magna Graecia" University of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences Viale S. Venuta 88100 Catanzaro Italy
| | - Massimo Fresta
- Department of Experimental and Clinical Medicine, Research Center "ProHealth Translational Hub", "Magna Graecia" University of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences Viale S. Venuta 88100 Catanzaro Italy
- Department of Health Sciences, University of Catanzaro "Magna Graecia" Viale "S. Venuta" 88100 Catanzaro Italy
| | - Stefania Orrù
- CEINGE-Biotecnologie Avanzate Franco Salvatore Naples 80145 Italy +39 081 3737880
- Department of Medical, Movement and Wellness Sciences, University of Naples Parthenope Naples 80133 Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio" Via dei Vestini 31 66100 Chieti Italy +39 0871 3554711
- Lithuanian University of Health Sciences, Laboratory of Drug Targets Histopathology, Institute of Cardiology A. Mickeviciaus g. 9 LT-44307 Kaunas Lithuania
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University Shanghai 200444 China
- UdA-TechLab, Research Center, University of Chieti-Pescara "G. D'Annunzio" 66100 Chieti Italy
| | - Francesco Salvatore
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II Naples 80131 Italy +39 3356069177
- CEINGE-Biotecnologie Avanzate Franco Salvatore Naples 80145 Italy +39 081 3737880
| |
Collapse
|
4
|
Cavallaro PA, De Santo M, Belsito EL, Longobucco C, Curcio M, Morelli C, Pasqua L, Leggio A. Peptides Targeting HER2-Positive Breast Cancer Cells and Applications in Tumor Imaging and Delivery of Chemotherapeutics. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2476. [PMID: 37686984 PMCID: PMC10490457 DOI: 10.3390/nano13172476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023]
Abstract
Breast cancer represents the most common cancer type and one of the major leading causes of death in the female worldwide population. Overexpression of HER2, a transmembrane glycoprotein related to the epidermal growth factor receptor, results in a biologically and clinically aggressive breast cancer subtype. It is also the primary driver for tumor detection and progression and, in addition to being an important prognostic factor in women diagnosed with breast cancer, HER2 is a widely known therapeutic target for drug development. The aim of this review is to provide an updated overview of the main approaches for the diagnosis and treatment of HER2-positive breast cancer proposed in the literature over the past decade. We focused on the different targeting strategies involving antibodies and peptides that have been explored with their relative outcomes and current limitations that need to be improved. The review also encompasses a discussion on targeted peptides acting as probes for molecular imaging. By using different types of HER2-targeting strategies, nanotechnology promises to overcome some of the current clinical challenges by developing novel HER2-guided nanosystems suitable as powerful tools in breast cancer imaging, targeting, and therapy.
Collapse
Affiliation(s)
- Palmira Alessia Cavallaro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Marzia De Santo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Emilia Lucia Belsito
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Camilla Longobucco
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Manuela Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Catia Morelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Luigi Pasqua
- Department of Environmental Engineering, University of Calabria, Via P. Bucci, 87036 Rende, Italy
| | - Antonella Leggio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| |
Collapse
|
5
|
Shafaei N, Khorshidi S, Karkhaneh A. The immune-stealth polymeric coating on drug delivery nanocarriers: In vitro engineering and in vivo fate. J Biomater Appl 2023:8853282231185352. [PMID: 37480331 DOI: 10.1177/08853282231185352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Although essential nanosystems such as nanoparticles and nanocarriers are desirable options for transporting various drug molecules into the biological environment, they rapidly remove from the circulatory system due to their interaction with multiple in vivo barriers, especially the immune barrier, which will result in their short-term effects. In order to improve their effectiveness and durability in the circulatory system, the polymer coatings can use to cover the surface of nanoparticles and nanocarriers to conceal them from the immune system. Due to their different properties (like charge, elasticity, and hydrophilicity/hydrophobicity), these coatings can improve drug delivery nanosystem durability and therapeutic applications. The mentioned coatings have different types and are divided into various categories, such as synthetic polymers, polysaccharides, and zwitterionic polymers. Each of these polymers has unique properties based on its category, origin, and chemical structure that make them suitable for producing stealth drug delivery nanocarriers. In this review article, we have tried to explain the importance of these diverse polymer coatings in determining the fate of drug nanocarriers and then introduced the different types of these coatings and, finally, described various methods that directly and indirectly analyze the nanocoatings to determine the stability of nanoparticles in the body.
Collapse
Affiliation(s)
- Nadia Shafaei
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Sajedeh Khorshidi
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Akbar Karkhaneh
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
6
|
Lim C, Shin Y, Lee S, Lee S, Lee MY, Shin BS, Oh KT. Dynamic drug release state and PEG length in PEGylated liposomal formulations define the distribution and pharmacological performance of drug. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
7
|
Glassman PM, Villa CH, Marcos-Contreras OA, Hood ED, Walsh LR, Greineder CF, Myerson JW, Shuvaeva T, Puentes L, Brenner JS, Siegel DL, Muzykantov VR. Targeted In Vivo Loading of Red Blood Cells Markedly Prolongs Nanocarrier Circulation. Bioconjug Chem 2022; 33:1286-1294. [PMID: 35710322 DOI: 10.1021/acs.bioconjchem.2c00196] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Engineering drug delivery systems for prolonged pharmacokinetics (PK) has been an ongoing pursuit for nearly 50 years. The gold standard for PK enhancement is the coating of nanoparticles with polymers, namely polyethylene glycol (PEGylation), which has been applied in several clinically used products. In the present work, we utilize the longest circulating and most abundant component of blood─the erythrocyte─to improve the PK behavior of liposomes. Antibody-mediated coupling of liposomes to erythrocytes was tested in vitro to identify a loading dose that did not adversely impact the carrier cells. Injection of erythrocyte targeting liposomes into mice resulted in a ∼2-fold improvement in the area under the blood concentration versus time profile versus PEGylated liposomes and a redistribution from the plasma into the cellular fraction of blood. These results suggest that in vivo targeting of erythrocytes is a viable strategy to improve liposome PK relative to current, clinically viable strategies.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Carlos H Villa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Landis R Walsh
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Colin F Greineder
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Laura Puentes
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Don L Siegel
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
8
|
Nsairat H, Khater D, Sayed U, Odeh F, Al Bawab A, Alshaer W. Liposomes: structure, composition, types, and clinical applications. Heliyon 2022; 8:e09394. [PMID: 35600452 PMCID: PMC9118483 DOI: 10.1016/j.heliyon.2022.e09394] [Citation(s) in RCA: 414] [Impact Index Per Article: 138.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/19/2022] [Accepted: 05/06/2022] [Indexed: 12/18/2022] Open
Abstract
Liposomes are now considered the most commonly used nanocarriers for various potentially active hydrophobic and hydrophilic molecules due to their high biocompatibility, biodegradability, and low immunogenicity. Liposomes also proved to enhance drug solubility and controlled distribution, as well as their capacity for surface modifications for targeted, prolonged, and sustained release. Based on the composition, liposomes can be considered to have evolved from conventional, long-circulating, targeted, and immune-liposomes to stimuli-responsive and actively targeted liposomes. Many liposomal-based drug delivery systems are currently clinically approved to treat several diseases, such as cancer, fungal and viral infections; more liposomes have reached advanced phases in clinical trials. This review describes liposomes structure, composition, preparation methods, and clinical applications.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Dima Khater
- Department of Chemistry, Faculty of Arts and Science, Applied Science Private University, Amman, Jordan
| | - Usama Sayed
- Department of Biology, The University of Jordan, Amman, 11942, Jordan
| | - Fadwa Odeh
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan
| | - Abeer Al Bawab
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.,Hamdi Mango Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
9
|
Münter R, Stavnsbjerg C, Christensen E, Thomsen ME, Stensballe A, Hansen AE, Parhamifar L, Kristensen K, Simonsen JB, Larsen JB, Andresen TL. Unravelling Heterogeneities in Complement and Antibody Opsonization of Individual Liposomes as a Function of Surface Architecture. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106529. [PMID: 35187804 DOI: 10.1002/smll.202106529] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/24/2022] [Indexed: 06/14/2023]
Abstract
Coating nanoparticles with poly(ethylene glycol) (PEG) is widely used to achieve long-circulating properties after infusion. While PEG reduces binding of opsonins to the particle surface, immunogenic anti-PEG side-effects show that PEGylated nanoparticles are not truly "stealth" to surface active proteins. A major obstacle for understanding the complex interplay between opsonins and nanoparticles is the averaging effects of the bulk assays that are typically applied to study protein adsorption to nanoparticles. Here, a microscopy-based method for directly quantifying opsonization at the single nanoparticle level is presented. Various surface coatings are investigated on liposomes, including PEG, and show that opsonization by both antibodies and complement C3b is highly dependent on the surface chemistry. It is further demonstrated that this opsonization is heterogeneous, with opsonized and non-opsonized liposomes co-existing in the same ensemble. Surface coatings modify the percentage of opsonized liposomes and/or opsonin surface density on the liposomes, with strikingly different patterns for antibodies and complement. Thus, this assay provides mechanistic details about opsonization at the single nanoparticle level previously inaccessible to established bulk assays.
Collapse
Affiliation(s)
- Rasmus Münter
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Camilla Stavnsbjerg
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Esben Christensen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Mikkel E Thomsen
- Department of Health Science and Technology, Aalborg University, Aalborg Ø, 9220, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg Ø, 9220, Denmark
| | - Anders E Hansen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Ladan Parhamifar
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Kasper Kristensen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Jens B Simonsen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Jannik B Larsen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| | - Thomas L Andresen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, 2800, Denmark
| |
Collapse
|
10
|
van der Koog L, Gandek TB, Nagelkerke A. Liposomes and Extracellular Vesicles as Drug Delivery Systems: A Comparison of Composition, Pharmacokinetics, and Functionalization. Adv Healthc Mater 2022; 11:e2100639. [PMID: 34165909 PMCID: PMC11468589 DOI: 10.1002/adhm.202100639] [Citation(s) in RCA: 169] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/27/2021] [Indexed: 12/11/2022]
Abstract
Over the past decades, lipid-based nanoparticle drug delivery systems (DDS) have caught the attention of researchers worldwide, encouraging the field to rapidly develop improved ways for effective drug delivery. One of the most prominent examples is liposomes, which are spherical shaped artificial vesicles composed of lipid bilayers and able to encapsulate both hydrophilic and hydrophobic materials. At the same time, biological nanoparticles naturally secreted by cells, called extracellular vesicles (EVs), have emerged as promising more complex biocompatible DDS. In this review paper, the differences and similarities in the composition of both vesicles are evaluated, and critical mediators that affect their pharmacokinetics are elucidate. Different strategies that have been assessed to tweak the pharmacokinetics of both liposomes and EVs are explored, detailing the effects on circulation time, targeting capacity, and cytoplasmic delivery of therapeutic cargo. Finally, whether a hybrid system, consisting of a combination of only the critical constituents of both vesicles, could offer the best of both worlds is discussed. Through these topics, novel leads for further research are provided and, more importantly, gain insight in what the liposome field and the EV field can learn from each other.
Collapse
Affiliation(s)
- Luke van der Koog
- Molecular PharmacologyGroningen Research Institute of PharmacyGRIAC Research Institute, University Medical Center GroningenUniversity of GroningenP.O. Box 196, XB10Groningen9700 ADThe Netherlands
| | - Timea B. Gandek
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| | - Anika Nagelkerke
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| |
Collapse
|
11
|
Zhang W, Callmann CE, Mirkin CA. Controlling the Biological Fate of Liposomal Spherical Nucleic Acids Using Tunable Polyethylene Glycol Shells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:46325-46333. [PMID: 34547202 PMCID: PMC8590845 DOI: 10.1021/acsami.1c12852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Liposomal spherical nucleic acids (LSNAs) modified with polyethylene glycol (PEG) units are studied in an attempt to understand how the circulation time and biodistribution of the constructs can be manipulated. Specifically, the effect of (1) PEG molecular weight, (2) PEG shell stability, and (3) PEG modification method (PEG in both the core and shell versus PEG in the shell only) on LSNA blood circulation, biodistribution, and in vivo cell internalization in a syngeneic, orthotopic triple-negative breast cancer mouse model is studied. Generally, high PEG molecular weight extends blood circulation lifetime, and a more lipophilic anchor stabilizes the PEG shell and improves circulation and tumor accumulation but at the cost of cell uptake efficiency. The PEGylation strategy has a minor effect on in vitro properties of LSNAs but significantly alters in vivo cell uptake. For example, surface-only PEG in one design contributed to higher in vivo cell internalization than its counterpart with PEG both in the shell and core. Taken together, this work provides guidelines for designing LSNAs that exhibit maximal in vivo cancer cell uptake characteristics in the context of a breast cancer model.
Collapse
Affiliation(s)
- Wuliang Zhang
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Cassandra E Callmann
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A Mirkin
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
12
|
Mihalik NE, Wen S, Driesschaert B, Eubank TD. Formulation and In Vitro Characterization of PLGA/PLGA-PEG Nanoparticles Loaded with Murine Granulocyte-Macrophage Colony-Stimulating Factor. AAPS PharmSciTech 2021; 22:191. [PMID: 34169366 DOI: 10.1208/s12249-021-02049-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) has demonstrated notable clinical activity in cancer immunotherapy, but it is limited by systemic toxicities, poor bioavailability, rapid clearance, and instability in vivo. Nanoparticles (NPs) may overcome these limitations and provide a mechanism for passive targeting of tumors. This study aimed to develop GM-CSF-loaded PLGA/PLGA-PEG NPs and evaluate them in vitro as a potential candidate for in vivo administration. NPs were created by a phase-separation technique that did not require toxic/protein-denaturing solvents or harsh agitation techniques and encapsulated GM-CSF in a more stable precipitated form. NP sizes were within 200 nm for enhanced permeability and retention (EPR) effect with negative zeta potentials, spherical morphology, and high entrapment efficiencies. The optimal formulation was identified by sustained release of approximately 70% of loaded GM-CSF over 24 h, alongside an average size of 143 ± 35 nm and entrapment efficiency of 84 ± 5%. These NPs were successfully freeze-dried in 5% (w/v) hydroxypropyl-β-cyclodextrin for long-term storage and further characterized. Bioactivity of released GM-CSF was determined by observing GM-CSF receptor activation on murine monocytes and remained fully intact. NPs were not cytotoxic to murine bone marrow-derived macrophages (BMDMs) at concentrations up to 1 mg/mL as determined by MTT and trypan blue exclusion assays. Lastly, NP components generated no significant transcription of inflammation-regulating genes from BMDMs compared to IFNγ+LPS "M1" controls. This report lays the preliminary groundwork to validate in vivo studies with GM-CSF-loaded PLGA/PEG-PLGA NPs for tumor immunomodulation. Overall, these data suggest that in vivo delivery will be well tolerated.
Collapse
|
13
|
Rommasi F, Esfandiari N. Liposomal Nanomedicine: Applications for Drug Delivery in Cancer Therapy. NANOSCALE RESEARCH LETTERS 2021; 16:95. [PMID: 34032937 PMCID: PMC8149564 DOI: 10.1186/s11671-021-03553-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/17/2021] [Indexed: 05/23/2023]
Abstract
The increasing prevalence of cancer, a disease in which rapid and uncontrollable cell growth causes complication and tissue dysfunction, is one of the serious and tense concerns of scientists and physicians. Nowadays, cancer diagnosis and especially its effective treatment have been considered as one of the biggest challenges in health and medicine in the last century. Despite significant advances in drug discovery and delivery, their many adverse effects and inadequate specificity and sensitivity, which usually cause damage to healthy tissues and organs, have been great barriers in using them. Limitation in the duration and amount of these therapeutic agents' administration is also challenging. On the other hand, the incidence of tumor cells that are resistant to typical methods of cancer treatment, such as chemotherapy and radiotherapy, highlights the intense need for innovation, improvement, and development in antitumor drug properties. Liposomes have been suggested as a suitable candidate for drug delivery and cancer treatment in nanomedicine due to their ability to store drugs with different physical and chemical characteristics. Moreover, the high flexibility and potential of liposome structure for chemical modification by conjugating various polymers, ligands, and molecules is a significant pro for liposomes not only to enhance their pharmacological merits but also to improve the effectiveness of anticancer drugs. Liposomes can increase the sensitivity, specificity, and durability of these anti-malignant cell agents in the body and provide remarkable benefits to be applied in nanomedicines. We reviewed the discovery and development of liposomes focusing on their clinical applications to treat diverse sorts of cancers and diseases. How the properties of liposomal drugs can be improved and their opportunity and challenges for cancer therapy were also considered and discussed.
Collapse
Affiliation(s)
- Foad Rommasi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Neda Esfandiari
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
14
|
Liposomes Prevent In Vitro Hemolysis Induced by Streptolysin O and Lysenin. MEMBRANES 2021; 11:membranes11050364. [PMID: 34069894 PMCID: PMC8157566 DOI: 10.3390/membranes11050364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/08/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022]
Abstract
The need for alternatives to antibiotics in the fight against infectious diseases has inspired scientists to focus on antivirulence factors instead of the microorganisms themselves. In this respect, prior work indicates that tiny, enclosed bilayer lipid membranes (liposomes) have the potential to compete with cellular targets for toxin binding, hence preventing their biological attack and aiding with their clearance. The effectiveness of liposomes as decoy targets depends on their availability in the host and how rapidly they are cleared from the circulation. Although liposome PEGylation may improve their circulation time, little is known about how such a modification influences their interactions with antivirulence factors. To fill this gap in knowledge, we investigated regular and long-circulating liposomes for their ability to prevent in vitro red blood cell hemolysis induced by two potent lytic toxins, lysenin and streptolysin O. Our explorations indicate that both regular and long-circulating liposomes are capable of similarly preventing lysis induced by streptolysin O. In contrast, PEGylation reduced the effectiveness against lysenin-induced hemolysis and altered binding dynamics. These results suggest that toxin removal by long-circulating liposomes is feasible, yet dependent on the particular virulence factor under scrutiny.
Collapse
|
15
|
Song Y, She Z, Huang Z, Wang S, Liu X, Zhang Q, Sun J, Di D, Deng Y. Are third-generation active-targeting nanoformulations definitely the best? In vitro and in vivo comparisons of pixantrone-loaded liposomes modified with different sialic acid derivatives. Drug Deliv Transl Res 2021; 12:647-661. [PMID: 33928513 DOI: 10.1007/s13346-021-00973-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2021] [Indexed: 11/26/2022]
Abstract
Treatment with sialic acid-octadecylamine (SA-ODA)-modified pixantrone (Pix) liposomes results in favorable antitumor effects by targeting tumor-associated macrophages (TAMs). To explore the influence of different types of SA decorations on antitumor efficiency, we synthesized a PEGylated SA derivative, SA-PEG2000-DSPE, and combined it with SA-ODA to construct three representative types of SA-modified liposomes (SA-ODA-modified Pix liposomes, SA-ODA-modified Pix liposomes with different PEG densities, and SA-PEG2000-DSPE-modified Pix liposomes, named Pix-SACL, Pix-SPL-0.2/0.5/2.0/5.0, and Pix-SAPL, respectively). All the Pix liposomes were nanoscale formulations, having diameters between 100 and 150 nm, high encapsulation efficiencies (> 90%), and slow drug release properties. The in vivo blood circulation time of the PEGylated formulations (Pix-SPL-0.2/0.5/2.0/5.0 and Pix-SAPL) showed an upward trend with increasing PEG density, but there was no significant difference between adjacent groups. All PEGylated formulations displayed increased tumor accumulation when compared with Pix-SACL, but there was no significant difference among them. However, the antitumor activity of SA-modified liposomes was not positively correlated with circulation time or tumor accumulation in S180-bearing mice. Pix-SPL-0.2 displayed the strongest antitumor effect and lowest toxicity among the formulations tested in this study. With Pix-SPL-0.2 treatment, 66.7% of the mice demonstrated tumor shedding and wound healing.
Collapse
Affiliation(s)
- Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhennan She
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
- School of Pharmaceutical Science & Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China.
| | - Zhenjun Huang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Shuo Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Qi Zhang
- Department of General Surgery, General Hospital of Benxi Iron and Steel Co., Ltd, Benxi, China
| | - Jing Sun
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Donghua Di
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
16
|
Nandi U, Onyesom I, Douroumis D. Anti-cancer activity of sirolimus loaded liposomes in prostate cancer cell lines. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
17
|
Nordström R, Zhu L, Härmark J, Levi-Kalisman Y, Koren E, Barenholz Y, Levinton G, Shamrakov D. Quantitative Cryo-TEM Reveals New Structural Details of Doxil-Like PEGylated Liposomal Doxorubicin Formulation. Pharmaceutics 2021; 13:pharmaceutics13010123. [PMID: 33478023 PMCID: PMC7835975 DOI: 10.3390/pharmaceutics13010123] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/15/2021] [Accepted: 01/15/2021] [Indexed: 11/17/2022] Open
Abstract
Nano-drugs based on nanoparticles (NP) or on nano-assemblies as carriers of the active pharmaceutical ingredient (API) are often expected to perform better compared to conventional dosage forms. Maximum realization of this potential though requires optimization of multiple physico-chemical, including structural and morphological, parameters. Meaningful distributions of these parameters derived from sufficient populations of individual NPs rather than ensemble distributions are desirable for this task, provided that relevant high-resolution data is available. In this study we demonstrate powerful capabilities of the up-to-date cryogenic transmission electron-microscopy (cryo-TEM) as well as correlations with other techniques abundant in the nano-research milieu. We explored Doxil®-like (an anticancer drug and the first FDA-approved nano-drug) (75–100 nm) PEGylated liposomes encapsulating single doxorubicin-sulfate nano-rod-crystals (PLD). These crystals induce liposome sphere-to-ellipsoid deformation. Doxil® was characterized by a multitude of physicochemical methods. We demonstrate, that accompanied by advanced image-analysis means, cryo-TEM can successfully enable the determination of multiple structural parameters of such complex liposomal nano-drugs with an added value of statistically-sound distributions. The latter could not be achieved by most other physicochemical approaches. It seems that cryo-TEM is capable of quantitative description of individual liposome morphological features, including meaningful distributions of all structural elements, with averages that correlate with other physical methods. Here it is demonstrated that such quantitative cryo-TEM analysis is a powerful tool in determining what is the optimal drug to lipid ratio in PLD, which is found to be the drug to lipid ratio existing in Doxil®.
Collapse
Affiliation(s)
- Rickard Nordström
- Vironova AB, Gävlegatan 22, 113 30 Stockholm, Sweden; (R.N.); (L.Z.); (J.H.)
| | - Lin Zhu
- Vironova AB, Gävlegatan 22, 113 30 Stockholm, Sweden; (R.N.); (L.Z.); (J.H.)
| | - Johan Härmark
- Vironova AB, Gävlegatan 22, 113 30 Stockholm, Sweden; (R.N.); (L.Z.); (J.H.)
| | - Yael Levi-Kalisman
- Institute for Life Sciences and The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel;
| | - Erez Koren
- Laboratory of Membrane and Liposome Research, Hadassah Medical School, The Hebrew University, IMRIC, Jerusalem 9112001, Israel; (E.K.); (Y.B.)
| | - Yechezkel Barenholz
- Laboratory of Membrane and Liposome Research, Hadassah Medical School, The Hebrew University, IMRIC, Jerusalem 9112001, Israel; (E.K.); (Y.B.)
| | - Genia Levinton
- Ayana Pharma Ltd., Hadassah Ein Kerem Campus Biotechnology Park, Jerusalem 9112002, Israel;
| | - Dima Shamrakov
- Ayana Pharma Ltd., Hadassah Ein Kerem Campus Biotechnology Park, Jerusalem 9112002, Israel;
- Correspondence: ; Tel.: +972-55-666-0711
| |
Collapse
|
18
|
Jiao J, Jiao X, Wang C, Wei L, Wang G, Deng Y, Song Y. The Contribution of PEG Molecular Weights in PEGylated Emulsions to the Various Phases in the Accelerated Blood Clearance (ABC) Phenomenon in Rats. AAPS PharmSciTech 2020; 21:300. [PMID: 33140142 DOI: 10.1208/s12249-020-01838-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/30/2020] [Indexed: 11/30/2022] Open
Abstract
PEGylated preparations will be cleared rapidly from blood circulation when they are administrated twice in the same animal at a time interval, referred to as the "accelerated blood clearance" (ABC) phenomenon. Commonly, the study of the ABC phenomenon was investigated in two aspects: induction phase and effectuation phase. Herein, we report the influence of physicochemical properties (PEG molecular weights) in the induction phase and effectuation phase on the ABC phenomenon. In the experiment, on one hand, PEGylated emulsions with different molecular weights of PEG (refer to PEn, n = 400, 600, 800, 1000, 2000, and 5000) were injected for the first dose (induction phase) and induced PE2000 to produce ABC phenomenon. On the other hand, after PE2000 injected, PEn was injected for the second dose (effectuation phase). The results indicated that PE2000 and PE5000 induced an intense ABC phenomenon by their long-circulating characteristic. Interestingly, PE400, PE600, PE800, and PE1000 produced a consistent ABC phenomenon but different circulation time. Apparently, the induction of the ABC phenomenon is not only determined by the circulation time but also by the PEG molecular weights. When PEn is in the effectuation phase, the extent of the ABC phenomenon was not positively related to the molecular weights of PEG, increasing first and then weaken with the increase of molecular weights of PEG. These suggest that the number of -(CH2CH2O)n- repeat units of PE2000 was more conducive to interact with anti-PEG IgM. The results reported here clearly indicate that both the PEG molecular weights of prior dose and the subsequent dose of emulsion strongly influence the extent of the ABC phenomenon. Taken together, our observations in this study complete the effect of PEG molecular weights at a different phase of the ABC phenomenon. Furthermore, our findings have a significant impact on the choice of molecular weights for PEGylated formulations for use in cross-administration.
Collapse
|
19
|
Di J, Xie F, Xu Y. When liposomes met antibodies: Drug delivery and beyond. Adv Drug Deliv Rev 2020; 154-155:151-162. [PMID: 32926944 DOI: 10.1016/j.addr.2020.09.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022]
Abstract
Drug encapsulated liposomes and monoclonal antibodies (Mabs) are two distinctively different classes of therapeutics, but both aim to become the ultimate "magic bullet". While PEGylated liposomes rely on the enhanced permeability and retention (EPR) effect for accumulation in solid tumor tissues, Mabs are designed to bind tightly to specific surface antigens on target cells to exert effector functions. Immunoliposome (IL) refers to the structural combination of liposomes and antibodies, whereas the antibodies are usually decorated on the liposome surface. ILs can therefore take advantage of interactions between antibodies and cancer cells for more efficient endocytosis and intracellular drug delivery. The antibody structure, affinity, density, as well as the liposome surface properties and drug to lipid ratios all contribute to the IL pharmacokinetic(PK) and pharmacodynamic(PD) behaviors. The optimal formulation parameters may vary for different target cells and tissues. Furthermore, besides the delivery of cytotoxic drugs to cancer cells, new ILs are being developed to interact with multiple target receptors, multiple target cells and trigger multiple therapeutic effects. We envision that the IL format can be a great platform for the molecular engineering of multi-valent, multi-specific interactions to achieve complex biological functions for therapeutic benefits, especially in the area of cancer immunotherapy.
Collapse
Affiliation(s)
- Jiaxing Di
- School of Pharmacy, Shanghai Jiao Tong University, China
| | - Fang Xie
- Department of Biomedical Engineering, Johns Hopkins University, United States of America
| | - Yuhong Xu
- College of Pharmacy and Chemistry, Dali University, China.
| |
Collapse
|
20
|
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
|
21
|
Peng C, Huang Y, Zheng J. Renal clearable nanocarriers: Overcoming the physiological barriers for precise drug delivery and clearance. J Control Release 2020; 322:64-80. [PMID: 32194171 PMCID: PMC8696951 DOI: 10.1016/j.jconrel.2020.03.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/27/2020] [Accepted: 03/15/2020] [Indexed: 01/10/2023]
Abstract
Physiological barriers encountered in the clinical translation of cancer nanomedicines inspire the community to more deeply understand nano-bio interactions in not only tumor microenvironment but also entire body and develop new nanocarriers to tackle these barriers. Renal clearable nanocarriers are one kind of these newly emerged drug delivery systems (DDSs), which enable drugs to rapidly penetrate into the tumor cores with no need of long blood retention and escape macrophage uptake in the meantime they can also enhance body elimination of non-targeted anticancer drugs. As a result, they can improve therapeutic efficacies and reduce side effects of anticancer drugs. Not limited to anticancer drugs, diagnostic agents can also be achieved with these renal clearable DDSs, which might also be applied to improve the precision in the gene editing and immunotherapy in the future.
Collapse
Affiliation(s)
- Chuanqi Peng
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, USA
| | - Yingyu Huang
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, USA
| | - Jie Zheng
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, USA.
| |
Collapse
|
22
|
Simonsen JB, Münter R. Pay Attention to Biological Nanoparticles when Studying the Protein Corona on Nanomedicines. Angew Chem Int Ed Engl 2020; 59:12584-12588. [DOI: 10.1002/anie.202004611] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/17/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Jens B. Simonsen
- Department of Health Technology Technical University of Denmark 2800 Kgs. Lyngby Denmark
| | - Rasmus Münter
- Department of Health Technology Technical University of Denmark 2800 Kgs. Lyngby Denmark
| |
Collapse
|
23
|
Simonsen JB, Münter R. Pay Attention to Biological Nanoparticles when Studying the Protein Corona on Nanomedicines. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202004611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jens B. Simonsen
- Department of Health Technology Technical University of Denmark 2800 Kgs. Lyngby Denmark
| | - Rasmus Münter
- Department of Health Technology Technical University of Denmark 2800 Kgs. Lyngby Denmark
| |
Collapse
|
24
|
Fam SY, Chee CF, Yong CY, Ho KL, Mariatulqabtiah AR, Tan WS. Stealth Coating of Nanoparticles in Drug-Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E787. [PMID: 32325941 PMCID: PMC7221919 DOI: 10.3390/nano10040787] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 01/01/2023]
Abstract
Nanoparticles (NPs) have emerged as a powerful drug-delivery tool for cancer therapies to enhance the specificity of drug actions, while reducing the systemic side effects. Nonetheless, NPs interact massively with the surrounding physiological environments including plasma proteins upon administration into the bloodstream. Consequently, they are rapidly cleared from the blood circulation by the mononuclear phagocyte system (MPS) or complement system, resulting in a premature elimination that will cause the drug release at off-target sites. By grafting a stealth coating layer onto the surface of NPs, the blood circulation half-life of nanomaterials can be improved by escaping the recognition and clearance of the immune system. This review focuses on the basic concept underlying the stealth behavior of NPs by polymer coating, whereby the fundamental surface coating characteristics such as molecular weight, surface chain density as well as conformations of polymer chains are of utmost importance for efficient protection of NPs. In addition, the most commonly used stealth polymers such as poly(ethylene glycol) (PEG), poly(2-oxazoline) (POx), and poly(zwitterions) in developing long-circulating NPs for drug delivery are also thoroughly discussed. The biomimetic strategies, including the cell-membrane camouflaging technique and CD47 functionalization for the development of stealth nano-delivery systems, are highlighted in this review as well.
Collapse
Affiliation(s)
- See Yee Fam
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.Y.F.); (C.Y.Y.)
| | - Chin Fei Chee
- Nanotechnology and Catalysis Research Centre, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Chean Yeah Yong
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.Y.F.); (C.Y.Y.)
| | - Kok Lian Ho
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Abdul Razak Mariatulqabtiah
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
- Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Wen Siang Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.Y.F.); (C.Y.Y.)
- Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| |
Collapse
|
25
|
Garofalo M, Grazioso G, Cavalli A, Sgrignani J. How Computational Chemistry and Drug Delivery Techniques Can Support the Development of New Anticancer Drugs. Molecules 2020; 25:E1756. [PMID: 32290224 PMCID: PMC7180704 DOI: 10.3390/molecules25071756] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 01/17/2023] Open
Abstract
The early and late development of new anticancer drugs, small molecules or peptides can be slowed down by some issues such as poor selectivity for the target or poor ADME properties. Computer-aided drug design (CADD) and target drug delivery (TDD) techniques, although apparently far from each other, are two research fields that can give a significant contribution to overcome these problems. Their combination may provide mechanistic understanding resulting in a synergy that makes possible the rational design of novel anticancer based therapies. Herein, we aim to discuss selected applications, some also from our research experience, in the fields of anticancer small organic drugs and peptides.
Collapse
Affiliation(s)
- Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Giovanni Grazioso
- Department of Pharmaceutical Sciences, University of Milano, 20133 Milan, Italy
| | - Andrea Cavalli
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500 Bellinzona, Switzerland
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500 Bellinzona, Switzerland
| |
Collapse
|
26
|
Jiao J, Luo X, Wang C, Jiao X, Liu M, Liu X, Wei L, Deng Y, Song Y. Effects of Uncleavable and Cleavable PEG-Lipids with Different Molecular Weights on Accelerated Blood Clearance of PEGylated Emulsions in Beagle Dogs. AAPS PharmSciTech 2020; 21:106. [PMID: 32185548 DOI: 10.1208/s12249-020-1640-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/12/2020] [Indexed: 11/30/2022] Open
Abstract
To investigate the effect of polyethylene glycol (PEG) molecular weights on circulation time of PEGylated emulsions and the second injection of injected PEGylated emulsions, we studied the effect of molecular weights on the pharmacokinetic behavior of PEG-DSPE (modified emulsions with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-n-[methoxy (polyethyleneglycol)]) and PEG-CHMC (modified emulsions with poly(ethyleneglycol)-cholesteryl carbonate) emulsions in beagle dogs. The "accelerated blood clearance" (ABC) phenomenon was induced. Through this study, the contribution of PEG molecular weights on the ABC phenomenon was further clarified, and the results provided guidance for lessening or eliminating the ABC phenomenon. We injected different PEG-modified emulsions with 10% PEG-modified density into beagle dogs at 2 μmol phospholipids kg-1 and the blood samples were drawn after 1 min, 3 min, 5 min, 10 min, 15 min, 30 min, 60 min, 120 min, 240 min, 360 min, 600 min, and 24 h. Then, concentrations of the drug were assayed using high-performance liquid chromatography (HPLC). The results showed that the circulation times of PEG-DSPE-modified emulsions were significantly different because of the difference in molecular weights, whereas those of the PEG-CHMC modified emulsions were not. The spatial conformation of PEG with small molecular weights (PEG400, PEG600, and PEG800) was more likely to induce a strong ABC phenomenon. The results of our work suggest the interaction of circulation time and PEG molecular weights on the ABC phenomenon, implying that the spatial conformation of PEG has advantages that alleviate the ABC phenomenon. Importantly, the results have implications for the choice of molecular weights of PEG for PEGylated formulations.
Collapse
|
27
|
Yan J, Yin M, Foster FS, Démoré CEM. Tumor Contrast Imaging with Gas Vesicles by Circumventing the Reticuloendothelial System. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:359-368. [PMID: 31708270 DOI: 10.1016/j.ultrasmedbio.2019.09.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/07/2019] [Accepted: 09/17/2019] [Indexed: 06/10/2023]
Abstract
Gas vesicles (GVs) are nanosized structures (45-800 nm) and have been reported to produce non-linear contrast signals, making them an attractive agent for molecular targeting of tumors. One barrier to their use for pre-clinical oncology studies is rapid uptake into the reticuloendothelial system (RES) and consequent rapid decrease in contrast signal after infusion ends and low signal on reperfusion after a bubble burst sequence. The purpose of this study was to examine suppression of the RES and surface modification of GVs to prolong contrast circulation in tumors for ultrasound imaging. Ultrasound imaging to measure dynamics of contrast signal intensity in tumor models was carried out using a 21-MHz high-frequency array transducer with the Vevo 2100 ultrasound system. The non-linear contrast signal from intravenously injected GVs compared with peak enhancement was measured during contrast wash-out and on reperfusion after a contrast burst sequence. Disrupting the RES by saturating the macrophage population or chemically inhibiting the Kupffer cell population with gadolinium or Intralipid preserves 62%-88% of GVs' contrast enhancement relative to peak during the wash-out phase and 32%-56% on reperfusion compared with 38% and 14%, respectively, for no disruption of the RES, indicating longer circulation of GVs in the tumor. Additionally, coating the GVs with 2-, 5- or 10-kDa polyethylene glycol (PEG) chains resulted in >70% contrast signal retention in the tumors during wash-out and, for 5- or 10-kDa PEG chains, a return to >45% of peak contrast signal on reperfusion. These findings indicate that GVs can be used as a contrast agent for tumor imaging and that disruption of the RES improved recirculation and maintained contrast enhancement caused by GVs in the tumors.
Collapse
Affiliation(s)
- Judy Yan
- Sunnybrook Research Institute, Toronto, Ontario, Canada.
| | - Melissa Yin
- FUJIFILM VisualSonics Inc., Toronto, Ontario, Canada
| | - F Stuart Foster
- Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Christine E M Démoré
- Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
|
29
|
Abstract
Bioavailability is an ancient but effective terminology by which the entire therapeutic efficacy of a drug directly or indirectly relays. Despite considering general plasma bioavailability, specific organ/tissue bioavailability will pave the path to broad spectrum dose calculation. Clear knowledge and calculative vision on bioavailability can improve the research and organ-targeting phenomenon. This article comprises a detailed introduction on bioavailability along with regulatory aspects, kinetic data and novel bioformulative approaches to achieve improved organ specific bioavailability, which may not be readily related to blood plasma bioavailability.
Collapse
|
30
|
Partial Surface Modification of Low Generation Polyamidoamine Dendrimers: Gaining Insight into their Potential for Improved Carboplatin Delivery. Biomolecules 2019; 9:biom9060214. [PMID: 31159469 PMCID: PMC6627870 DOI: 10.3390/biom9060214] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 05/22/2019] [Accepted: 05/27/2019] [Indexed: 01/28/2023] Open
Abstract
Carboplatin (CAR) is a second generation platinum-based compound emerging as one of the most widely used anticancer drugs to treat a variety of tumors. In an attempt to address its dose-limiting toxicity and fast renal clearance, several delivery systems (DDSs) have been developed for CAR. However, unsuitable size range and low loading capacity may limit their potential applications. In this study, PAMAM G3.0 dendrimer was prepared and partially surface modified with methoxypolyethylene glycol (mPEG) for the delivery of CAR. The CAR/PAMAM G3.0@mPEG was successfully obtained with a desirable size range and high entrapment efficiency, improving the limitations of previous CAR-loaded DDSs. Cytocompatibility of PAMAM G3.0@mPEG was also examined, indicating that the system could be safely used. Notably, an in vitro release test and cell viability assays against HeLa, A549, and MCF7 cell lines indicated that CAR/PAMAM G3.0@mPEG could provide a sustained release of CAR while fully retaining its bioactivity to suppress the proliferation of cancer cells. These obtained results provide insights into the potential of PAMAM G3.0@mPEG dendrimer as an efficient delivery system for the delivery of a drug that has strong side effects and fast renal clearance like CAR, which could be a promising approach for cancer treatment.
Collapse
|
31
|
Nosova AS, Koloskova OO, Nikonova AA, Simonova VA, Smirnov VV, Kudlay D, Khaitov MR. Diversity of PEGylation methods of liposomes and their influence on RNA delivery. MEDCHEMCOMM 2019; 10:369-377. [PMID: 31015904 PMCID: PMC6457174 DOI: 10.1039/c8md00515j] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/10/2019] [Indexed: 12/18/2022]
Abstract
Gene therapy is a promising approach for personalized medicine, but its application in humans requires development of efficient and safe vehicles. PEGylated liposomes are some of the most suitable delivery systems for nucleic acids because of their stability under physiological conditions and prolonged circulation time, compared to conventional and other types of "stealth" liposomes. In vitro/in vivo activity of PEGylated liposomes is highly dependent on PEG motif abundance. The process of "stealth" coverage formation is a very important parameter for efficient transfection assays and further fate determination of the PEG layer after tissue penetration. In this review, we discuss the latest methods of PEGylated liposome preparation.
Collapse
Affiliation(s)
- A S Nosova
- NRC Institute of Immunology FMBA of Russia , Moscow , Russia .
| | - O O Koloskova
- NRC Institute of Immunology FMBA of Russia , Moscow , Russia .
| | - A A Nikonova
- NRC Institute of Immunology FMBA of Russia , Moscow , Russia .
- Mechnikov Research Institute of Vaccines and Sera , Moscow , Russia
| | - V A Simonova
- I. M. Sechenov First Moscow State Medical University , Moscow , Russia
| | - V V Smirnov
- NRC Institute of Immunology FMBA of Russia , Moscow , Russia .
- I. M. Sechenov First Moscow State Medical University , Moscow , Russia
| | - D Kudlay
- NRC Institute of Immunology FMBA of Russia , Moscow , Russia .
| | - M R Khaitov
- NRC Institute of Immunology FMBA of Russia , Moscow , Russia .
| |
Collapse
|
32
|
Luan X, Rahme K, Cong Z, Wang L, Zou Y, He Y, Yang H, Holmes JD, O'Driscoll CM, Guo J. Anisamide-targeted PEGylated gold nanoparticles designed to target prostate cancer mediate: Enhanced systemic exposure of siRNA, tumour growth suppression and a synergistic therapeutic response in combination with paclitaxel in mice. Eur J Pharm Biopharm 2019; 137:56-67. [PMID: 30779980 DOI: 10.1016/j.ejpb.2019.02.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 12/18/2018] [Accepted: 02/15/2019] [Indexed: 12/24/2022]
Abstract
Small interfering RNA (siRNA) has recently illustrated therapeutic potential for malignant disorders. However, the clinical application of siRNA-based therapeutics is significantly retarded by the paucity of successful delivery systems. Recently, multifunctional gold nanoparticles (AuNPs) as non-viral delivery carriers have shown promise for transporting chemotherapeutics, proteins/peptides, and genes. In this study, AuNPs capped with polyethylenimine (PEI) and PEGylated anisamide (a ligand known to target the sigma receptor) have been developed to produce a range of positively charged anisamide-targeted PEGylated AuNPs (namely Au-PEI-PEG-AA). The anisamide-targeted AuNPs effectively complexed siRNA via electrostatic interaction, and the resultant complex (Au110-PEI-PEG5000-AA.siRNA) illustrated favourable physicochemical characteristics, including particle size, surface charge, and stability. In vitro, anisamide-targeted AuNPs selectively bound to human prostate cancer PC-3 cells, inducing efficient endosomal escape of siRNA, and effective downregulation of the RelA gene. In vivo, prolonged systemic exposure of siRNA was achieved by anisamide-targeted AuNPs resulting in significant tumour growth suppression in a PC3 xenograft mouse model without an increase in toxicity. In addition, a combination of siRNA-mediated NF-κB knockdown using anisamide-targeted AuNPs with Paclitaxel produced a synergistic therapeutic response, thus providing a promising therapeutic strategy for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Xue Luan
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Kamil Rahme
- Department of Sciences, Faculty of Natural and Applied Science, Notre Dame University (Louaize), Zouk Mosbeh, Lebanon; Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland
| | - Zhongcheng Cong
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Limei Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Department of Pharmacy, The General Hospital of FAW, Changchun 130011, China
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yan He
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Hao Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Justin D Holmes
- Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland; AMBER@CRANN, Trinity College Dublin, Dublin 2, Ireland
| | | | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
33
|
Lynn GM, Chytil P, Francica JR, Lagová A, Kueberuwa G, Ishizuka AS, Zaidi N, Ramirez-Valdez RA, Blobel NJ, Baharom F, Leal J, Wang AQ, Gerner MY, Etrych T, Ulbrich K, Seymour LW, Seder RA, Laga R. Impact of Polymer-TLR-7/8 Agonist (Adjuvant) Morphology on the Potency and Mechanism of CD8 T Cell Induction. Biomacromolecules 2019; 20:854-870. [PMID: 30608149 DOI: 10.1021/acs.biomac.8b01473] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Small molecule Toll-like receptor-7 and -8 agonists (TLR-7/8a) can be used as vaccine adjuvants to induce CD8 T cell immunity but require formulations that prevent systemic toxicity and focus adjuvant activity in lymphoid tissues. Here, we covalently attached TLR-7/8a to polymers of varying composition, chain architecture and hydrodynamic behavior (∼300 nm submicrometer particles, ∼10 nm micelles and ∼4 nm flexible random coils) and evaluated how these parameters of polymer-TLR-7/8a conjugates impact adjuvant activity in vivo. Attachment of TLR-7/8a to any of the polymer compositions resulted in a nearly 10-fold reduction in systemic cytokines (toxicity). Moreover, both lymph node cytokine production and the magnitude of CD8 T cells induced against protein antigen increased with increasing polymer-TLR-7/8a hydrodynamic radius, with the submicrometer particle inducing the highest magnitude responses. Notably, CD8 T cell responses induced by polymer-TLR-7/8a were dependent on CCR2+ monocytes and IL-12, whereas responses by a small molecule TLR-7/8a that unexpectedly persisted in vaccine-site draining lymph nodes (T1/2 = 15 h) had less dependence on monocytes and IL-12 but required Type I IFNs. This study shows how modular properties of synthetic adjuvants can be chemically programmed to alter immunity in vivo through distinct immunological mechanisms.
Collapse
Affiliation(s)
- Geoffrey M Lynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Petr Chytil
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského nám. 2 , 162 06 Prague 6 , Czech Republic
| | - Joseph R Francica
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Anna Lagová
- Department of Oncology , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , United Kingdom
| | - Gray Kueberuwa
- Department of Oncology , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , United Kingdom
| | - Andrew S Ishizuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Neeha Zaidi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Ramiro A Ramirez-Valdez
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Nicolas J Blobel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Faezzah Baharom
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Joseph Leal
- Department of Immunology , University of Washington , South Lake Union E-411, 750 Republican Street , Seattle , Washington 98109 , United States
| | - Amy Q Wang
- Therapeutics for Rare and Neglected Diseases, National Center for Advancing Translational Sciences , 9800 Medical Center Drive , Rockville , Maryland 20850 , United States
| | - Michael Y Gerner
- Department of Immunology , University of Washington , South Lake Union E-411, 750 Republican Street , Seattle , Washington 98109 , United States
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského nám. 2 , 162 06 Prague 6 , Czech Republic
| | - Karel Ulbrich
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského nám. 2 , 162 06 Prague 6 , Czech Republic
| | - Leonard W Seymour
- Department of Oncology , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , United Kingdom
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Richard Laga
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského nám. 2 , 162 06 Prague 6 , Czech Republic
- Department of Oncology , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , United Kingdom
| |
Collapse
|
34
|
Nosova AS, Koloskova OO, Nikonova AA, Simonova VA, Smirnov VV, Kudlay D, Khaitov MR. Diversity of PEGylation methods of liposomes and their influence on RNA delivery. MEDCHEMCOMM 2019. [DOI: 10.1039/c8md00515j%0a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A brief review and comparison of the methods of PEGylation of liposomal particles and their influence on the delivery of RNA.
Collapse
Affiliation(s)
- A. S. Nosova
- NRC Institute of Immunology FMBA of Russia
- Moscow
- Russia
| | | | - A. A. Nikonova
- NRC Institute of Immunology FMBA of Russia
- Moscow
- Russia
- Mechnikov Research Institute of Vaccines and Sera
- Moscow
| | - V. A. Simonova
- I. M. Sechenov First Moscow State Medical University
- Moscow
- Russia
| | - V. V. Smirnov
- NRC Institute of Immunology FMBA of Russia
- Moscow
- Russia
- I. M. Sechenov First Moscow State Medical University
- Moscow
| | - D. Kudlay
- NRC Institute of Immunology FMBA of Russia
- Moscow
- Russia
| | - M. R. Khaitov
- NRC Institute of Immunology FMBA of Russia
- Moscow
- Russia
| |
Collapse
|
35
|
Maso K, Grigoletto A, Vicent MJ, Pasut G. Molecular platforms for targeted drug delivery. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 346:1-50. [DOI: 10.1016/bs.ircmb.2019.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
Malik P, Mukherjee TK. Recent advances in gold and silver nanoparticle based therapies for lung and breast cancers. Int J Pharm 2018; 553:483-509. [DOI: 10.1016/j.ijpharm.2018.10.048] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/20/2018] [Accepted: 10/20/2018] [Indexed: 02/06/2023]
|
37
|
Klein PM, Klinker K, Zhang W, Kern S, Kessel E, Wagner E, Barz M. Efficient Shielding of Polyplexes Using Heterotelechelic Polysarcosines. Polymers (Basel) 2018; 10:E689. [PMID: 30966723 PMCID: PMC6404158 DOI: 10.3390/polym10060689] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/13/2018] [Accepted: 06/17/2018] [Indexed: 11/16/2022] Open
Abstract
Shielding agents are commonly used to shield polyelectrolyte complexes, e.g., polyplexes, from agglomeration and precipitation in complex media like blood, and thus enhance their in vivo circulation times. Since up to now primarily poly(ethylene glycol) (PEG) has been investigated to shield non-viral carriers for systemic delivery, we report on the use of polysarcosine (pSar) as a potential alternative for steric stabilization. A redox-sensitive, cationizable lipo-oligomer structure (containing two cholanic acids attached via a bioreducible disulfide linker to an oligoaminoamide backbone in T-shape configuration) was equipped with azide-functionality by solid phase supported synthesis. After mixing with small interfering RNA (siRNA), lipopolyplexes formed spontaneously and were further surface-functionalized with polysarcosines. Polysarcosine was synthesized by living controlled ring-opening polymerization using an azide-reactive dibenzo-aza-cyclooctyne-amine as an initiator. The shielding ability of the resulting formulations was investigated with biophysical assays and by near-infrared fluorescence bioimaging in mice. The modification of ~100 nm lipopolyplexes was only slightly increased upon functionalization. Cellular uptake into cells was strongly reduced by the pSar shielding. Moreover, polysarcosine-shielded polyplexes showed enhanced blood circulation times in bioimaging studies compared to unshielded polyplexes and similar to PEG-shielded polyplexes. Therefore, polysarcosine is a promising alternative for the shielding of non-viral, lipo-cationic polyplexes.
Collapse
Affiliation(s)
- Philipp Michael Klein
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, Pharmaceutical Biotechnology, Butenandtstrasse 5-13, D-81377 Munich, Germany.
| | - Kristina Klinker
- Institute of Organic Chemistry, Johannes Gutenberg University, Duesbergweg 10-14, D-55128 Mainz, Germany.
- Graduate School Materials Science in Mainz, Staudinger Weg 9, 55128 Mainz, Germany.
| | - Wei Zhang
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, Pharmaceutical Biotechnology, Butenandtstrasse 5-13, D-81377 Munich, Germany.
| | - Sarah Kern
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, Pharmaceutical Biotechnology, Butenandtstrasse 5-13, D-81377 Munich, Germany.
| | - Eva Kessel
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, Pharmaceutical Biotechnology, Butenandtstrasse 5-13, D-81377 Munich, Germany.
| | - Ernst Wagner
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, Pharmaceutical Biotechnology, Butenandtstrasse 5-13, D-81377 Munich, Germany.
- Nanosystems Initiative Munich, Schellingstraße 4, D-80799 Munich, Germany.
| | - Matthias Barz
- Institute of Organic Chemistry, Johannes Gutenberg University, Duesbergweg 10-14, D-55128 Mainz, Germany.
| |
Collapse
|
38
|
Rodallec A, Fanciullino R, Lacarelle B, Ciccolini J. Seek and destroy: improving PK/PD profiles of anticancer agents with nanoparticles. Expert Rev Clin Pharmacol 2018; 11:599-610. [PMID: 29768060 DOI: 10.1080/17512433.2018.1477586] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The Pharmacokinetics/pharmacodynamics (PK/PD) relationships with cytotoxics are usually based on a steepening concentration-effect relationship; the greater the drug amount, the greater the effect. The Maximum Tolerated Dose paradigm, finding the balance between efficacy, while keeping toxicities at their manageable level, has been the rule of thumb for the last 50-years. Developing nanodrugs is an appealing strategy to help broaden this therapeutic window. The fact that efficacy and toxicity with cytotoxics are intricately linked is primarily due to the complete lack of specificity toward the tumor tissue during their distribution phase. Because nanoparticles are expected to better target tumor tissue while sparing healthy cells, accumulating large amounts of cytotoxics in tumors could be achieved in a safer way. Areas covered: This review aims at presenting how nanodrugs present unique features leading to reconsidering PK/PD relationships of anticancer agents. Expert commentary: The constant interplay between carrier PK, interactions with cancer cells, payload release, payload PK, target expression and target engagement, makes picturing the exact PK/PD relationships of nanodrugs particularly challenging. However, those improved PK/PD relationships now make the once contradictory higher efficacy and lower toxicities requirement an achievable goal in cancer patients.
Collapse
Affiliation(s)
- Anne Rodallec
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| | - Raphaelle Fanciullino
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| | - Bruno Lacarelle
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| | - Joseph Ciccolini
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| |
Collapse
|
39
|
Self-assembled amphiphilic core-shell nanocarriers in line with the modern strategies for brain delivery. J Control Release 2017. [PMID: 28648865 DOI: 10.1016/j.jconrel.2017.06.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Disorders of the central nervous system (CNS) represent increasing social and economic problems all over the world which makes the effective transport of drugs to the brain a crucial need. In the last decade, many strategies were introduced to deliver drugs to the brain trying to overcome the challenge of the blood brain barrier (BBB) using both invasive and non-invasive methods. Non-invasive strategy represented in the application of nanocarriers became very common. One of the most hopeful nanoscopic carriers for brain delivery is core-shell nanocarriers or polymeric micelles (PMs). They are more advantageous than other nanocarriers. They offer small size, ease of preparation, ease of sterilization and the possibility of surface modification with various ligands. Hence, the aim of this review is to discuss modern strategies for brain delivery, micelles as a successful delivery system for the brain and how micelles could be modified to act as "magic bullets" for brain delivery.
Collapse
|
40
|
Influence of Defined Hydrophilic Blocks within Oligoaminoamide Copolymers: Compaction versus Shielding of pDNA Nanoparticles. Polymers (Basel) 2017; 9:polym9040142. [PMID: 30970822 PMCID: PMC6432433 DOI: 10.3390/polym9040142] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/07/2017] [Accepted: 04/11/2017] [Indexed: 01/07/2023] Open
Abstract
Cationic polymers are promising components of the versatile platform of non-viral nucleic acid (NA) delivery agents. For a successful gene delivery system, these NA vehicles need to comprise several functionalities. This work focuses on the modification of oligoaminoamide carriers with hydrophilic oligomer blocks mediating nanoparticle shielding potential, which is necessary to prevent aggregation or dissociation of NA polyplexes in vitro, and hinder opsonization with blood components in vivo. Herein, the shielding agent polyethylene glycol (PEG) in three defined lengths (12, 24, or 48 oxyethylene repeats) is compared with two peptidic shielding blocks composed of four or eight repeats of sequential proline-alanine-serine (PAS). With both types of shielding agents, we found opposing effects of the length of hydrophilic segments on shielding and compaction of formed plasmid DNA (pDNA) nanoparticles. Two-arm oligoaminoamides with 37 cationizable nitrogens linked to 12 oxyethylene units or four PAS repeats resulted in very compact 40⁻50 nm pDNA nanoparticles, whereas longer shielding molecules destabilize the investigated polyplexes. Thus, the balance between sufficiently shielded but still compact and stable particles can be considered a critical optimization parameter for non-viral nucleic acid vehicles based on hydrophilic-cationic block oligomers.
Collapse
|
41
|
Safwat S, Ishak RA, Hathout RM, Mortada ND. Statins anticancer targeted delivery systems: re-purposing an old molecule. ACTA ACUST UNITED AC 2017; 69:613-624. [PMID: 28271498 DOI: 10.1111/jphp.12707] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/12/2017] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Exploring the use of statins as anticancer agents and exploiting different drug delivery systems in targeting these molecules to cancerous sites. Literature review was performed to investigate the use of statins in cancer treatment in one hand, and the different pharmaceutical approaches to deliver and target these drugs to their site of action. KEY FINDINGS Statins were used for decades as antihypercholestrolemic drugs but recently have been proven potential for broad anticancer activities. The incorporation of statins in nanoparticulate drug delivery systems not only augmented the cytotoxicity of statins but also overcame the resistance of cancerous cells against the traditional chemotherapeutic agents. Statins-loaded nanoparticles could be easily tampered to target the cancerous cells and consequently minimal drug amount could be utilized. SUMMARY This review reconnoitered the different endeavors to incorporate statins in various nanoparticles and summarized the successful effects in targeting cancerous cells and reducing their proliferation without the side effects of commonly used chemotherapeutic agents.
Collapse
Affiliation(s)
- Sally Safwat
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| | - Rania A Ishak
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| | - Rania M Hathout
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| | - Nahed D Mortada
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| |
Collapse
|
42
|
Guzman-Villanueva D, Migrino RQ, Truran S, Karamanova N, Franco DA, Burciu C, Senapati S, Nedelkov D, Hari P, Weissig V. PEGylated-nanoliposomal clusterin for amyloidogenic light chain-induced endothelial dysfunction. J Liposome Res 2017; 28:97-105. [PMID: 28103719 PMCID: PMC5591079 DOI: 10.1080/08982104.2016.1274756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Light chain (AL) amyloidosis is a disease associated with significant morbidity and mortality arising from multi-organ injury induced by amyloidogenic light chain proteins (LC). There is no available treatment to reverse the toxicity of LC. We previously showed that chaperone glycoprotein clusterin (CLU) and nanoliposomes (NL), separately, restore human microvascular endothelial function impaired by LC. In this work, we aim to prepare PEGylated-nanoliposomal clusterin (NL-CLU) formulations that could allow combined benefit against LC while potentially enabling efficient delivery to microvascular tissue, and test efficacy on human arteriole endothelial function. NL-CLU was prepared by a conjugation reaction between the carboxylated surface of NL and the primary amines of the CLU protein. NL were made of phosphatidylcholine (PC), cholesterol (Chol) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-2000] (DSPE-PEG 2000 carboxylic acid) at 70:25:5 mol%. The protective effect of NL-CLU was tested by measuring the dilation response to acetylcholine and papaverine in human adipose arterioles exposed to LC. LC treatment significantly reduced the dilation response to acetylcholine and papaverine; co-treatment of LC with PEGylated-nanoliposomal CLU or free CLU restored the dilator response. NL-CLU is a feasible and promising approach to reverse LC-induced endothelial damage.
Collapse
Affiliation(s)
- Diana Guzman-Villanueva
- a Department of Pharmaceutical Sciences , College of Pharmacy-Glendale, Midwestern University , AZ , USA.,b Nanomedicine Center of Excellence in Translation Cancer Research , Glendale , AZ , USA
| | - Raymond Q Migrino
- c Phoenix Veterans Affairs Health Care System , AZ , USA.,d University of Arizona College of Medicine-Phoenix , AZ , USA
| | - Seth Truran
- c Phoenix Veterans Affairs Health Care System , AZ , USA
| | | | | | - Camelia Burciu
- c Phoenix Veterans Affairs Health Care System , AZ , USA
| | - Subhadip Senapati
- e Department of Ophthalmology and Visual Sciences , Case Western Reserve University , OH , USA
| | - Dobrin Nedelkov
- f Biodesign Institute, Arizona State University , AZ , USA , and
| | - Parameswaran Hari
- g Department of Medicine , Medical College of Wisconsin , Milwaukee , WI , USA
| | - Volkmar Weissig
- a Department of Pharmaceutical Sciences , College of Pharmacy-Glendale, Midwestern University , AZ , USA.,b Nanomedicine Center of Excellence in Translation Cancer Research , Glendale , AZ , USA
| |
Collapse
|
43
|
Adumeau L, Genevois C, Roudier L, Schatz C, Couillaud F, Mornet S. Impact of surface grafting density of PEG macromolecules on dually fluorescent silica nanoparticles used for the in vivo imaging of subcutaneous tumors. Biochim Biophys Acta Gen Subj 2017; 1861:1587-1596. [PMID: 28179102 DOI: 10.1016/j.bbagen.2017.01.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND In the context of systematically administered nanomedicines, the physicochemistry of NP surfaces must be controlled as a prerequisite to improve blood circulation time, and passive and active targeting. In particular, there is a real need to develop NP stealth and labelling for both in vivo and microscopic fluorescence imaging in a mice model. METHODS We have synthesized NIR/red dually fluorescent silica nanoparticles of 19nm covalently covered by a PEG layer of different grafting density in the brush conformational regime by using a reductive amination reaction. These particles were characterized by TEM, DRIFT, DLS, TGA, ζ potential measurements, UV-vis and fluorescence spectroscopy. Prostate tumors were generated in mice by subcutaneous injection of RM1-CMV-Fluc cells. Tumor growth was monitored by BLI after a D-luciferin injection. Four samples of PEGylated fluorescent NPs were individually intravenously injected into 6 mice (N=6, total 24 mice). Nanoparticle distribution was investigated using in vivo fluorescence reflectance imaging (FRI) over 48h and microscopy imaging was employed to localize the NPs within tumors in vitro. RESULTS Fluorescent NP accumulation, due to the enhanced permeability and retention (EPR) effect, increases gradually as a function of increased PEG surface grafting density with a huge difference observed for the highest density grafting. For the highest grafting density, a blood circulation time of up to 24h was observed with a strong reduction in uptake by the liver. In vivo experimental results suggest that the biodistribution of NPs is very sensitive to slight variations in surface grafting density when the NPs present a high curvature radius. CONCLUSION This study underlines the need to compensate a high curvature radius with a PEG-saturated NP surface to improve blood circulation and accumulation within tumors through the EPR effect. Dually fluorescent NPs PEGylated to saturation display physical properties useful for assessing the susceptibility of tumors to the EPR effect. GENERAL SIGNIFICANCE Control of the physicochemical features of nanoparticle surfaces to improve blood circulation times and monitoring of the EPR effect. This article is part of a Special Issue entitled "Recent Advances in Bionanomaterials" Guest Editor: Dr. Marie-Louise Saboungi and Dr. Samuel D. Bader.
Collapse
Affiliation(s)
- Laurent Adumeau
- CNRS, Univ. Bordeaux, ICMCB, UPR 9048, F-33600 Pessac, France
| | - Coralie Genevois
- Univ. Bordeaux, EA 7435 IMOTION - Imagerie moléculaire et thérapies innovantes en oncologie, 33706 Bordeaux, France
| | - Lydia Roudier
- CNRS, Univ. Bordeaux, ICMCB, UPR 9048, F-33600 Pessac, France
| | | | - Franck Couillaud
- Univ. Bordeaux, EA 7435 IMOTION - Imagerie moléculaire et thérapies innovantes en oncologie, 33706 Bordeaux, France.
| | - Stéphane Mornet
- CNRS, Univ. Bordeaux, ICMCB, UPR 9048, F-33600 Pessac, France.
| |
Collapse
|
44
|
Structural analysis of binding functionality of folic acid-PEG dendrimers against folate receptor. J Mol Graph Model 2017; 72:201-208. [PMID: 28110184 DOI: 10.1016/j.jmgm.2017.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/23/2016] [Accepted: 01/04/2017] [Indexed: 12/27/2022]
Abstract
Dendrimers functionalized with folic acid (FA) are drug delivery systems that can selectively target cancer cells with folate receptors (FR-α) overexpression. Incorporation of polyethylene glycol (PEG) can enhance dendrimers solubility and pharmacokinetics, but ligand-receptor binding must not be affected. In this work we characterized, at atomic level, the binding functionality of conventional site-specific dendrimers conjugated with FA with PEG 750 or PEG 3350 as a linker. After Molecular Dynamics simulation, we observed that both PEG's did not interfere over ligand-receptor binding functionality. Although binding kinetics could be notably affected, the folate fragment from both dendrimers remained exposed to the solvent before approaching selectively to FR-α. PEG 3350 provided better solubility and protection from enzymatic degradation to the dendrimer than PEG 750. Also, FA-PEG3350 dendrimer showed a slightly better interaction with FR-α than FA-PEG750 dendrimer. Therefore, theoretical evidence supports that both dendrimers are suitable as drug delivery systems for cancer therapies.
Collapse
|
45
|
Zhang T, Zhou S, Hu L, Peng B, Liu Y, Luo X, Song Y, Liu X, Deng Y. Polysialic acid-modifying liposomes for efficient delivery of epirubicin, in-vitro characterization and in-vivo evaluation. Int J Pharm 2016; 515:449-459. [DOI: 10.1016/j.ijpharm.2016.10.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 10/06/2016] [Accepted: 10/23/2016] [Indexed: 12/22/2022]
|
46
|
Akhtar N, Khan RA. Liposomal systems as viable drug delivery technology for skin cancer sites with an outlook on lipid-based delivery vehicles and diagnostic imaging inputs for skin conditions'. Prog Lipid Res 2016; 64:192-230. [DOI: 10.1016/j.plipres.2016.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/15/2016] [Accepted: 08/09/2016] [Indexed: 12/19/2022]
|
47
|
Li H, Yuan D, Sun M, Ping Q. Effect of ligand density and PEG modification on octreotide-targeted liposome via somatostatin receptor in vitro and in vivo. Drug Deliv 2016; 23:3562-3572. [DOI: 10.1080/10717544.2016.1209797] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Huipeng Li
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Dongfen Yuan
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Qineng Ping
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
48
|
Molnar D, Linders J, Mayer C, Schubert R. Insertion stability of poly(ethylene glycol)-cholesteryl-based lipid anchors in liposome membranes. Eur J Pharm Biopharm 2016; 103:51-61. [DOI: 10.1016/j.ejpb.2016.03.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/15/2016] [Accepted: 03/21/2016] [Indexed: 10/22/2022]
|
49
|
Wei X, Cohen R, Barenholz Y. Insights into composition/structure/function relationships of Doxil® gained from "high-sensitivity" differential scanning calorimetry. Eur J Pharm Biopharm 2016; 104:260-70. [PMID: 27106607 DOI: 10.1016/j.ejpb.2016.04.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/18/2016] [Indexed: 12/11/2022]
Abstract
Thermotropic behavior of Doxil® and its generic, Lipodox®, was characterized using "high-sensitivity" differential scanning calorimetry (DSC). This is the first report that two distinct endotherms were observed in Doxil and Lipodox upon heating. The first (Tm at 51±2°C) is broad and of low enthalpy, representing the membrane lipid phase transition, which occurs despite having high (38mole%) cholesterol. The second (Tm at ∼70°C) is narrow, representing melting of the intraliposomal doxorubicin-sulfate nanocrystals. The thermograms of Doxil and Lipodox are practically identical. The membrane phase transition is similar to that of drug-free nanoliposomes of the same size and lipid composition as Doxil, suggesting lack of significant drug-membrane interaction. The melting endotherm of the intraliposomal nanocrystals is 2.0-2.5-fold narrower than that of the crystals formed in a solution of 250mM ammonium sulfate and >60mg/ml doxorubicin. This suggests that nanovolume of liposomes improves doxorubicin-sulfate crystallinity. Moreover, both phase transitions are reversible in cycled DSC scanning (15-90-15°C). This indicates an unexpected "non-leaky" phospholipid phase transition and excellent physical and chemical stabilities of Doxil after short exposure to high temperature. Reducing mole% of cholesterol results in a "leaky" membrane phase transition of higher enthalpy. Namely, high mole% cholesterol is essential for the resistance to drug leakage during phase transition. Pegylated liposomal doxorubicin in which HSPC was replaced by DPPC shows the same non-leaky phase transition but at a lower temperature, indicating this type of phase transition is not unique to Doxil. The presence of DSPE-PEG2k increases the cooperativity of the phase transition. High-sensitivity DSC helps illuminate composition/structure/function relationships of Doxil, and is useful for the equivalence/similarity studies.
Collapse
Affiliation(s)
- Xiaohui Wei
- Laboratory of Membrane and Liposome Research, The Hebrew University-Hadassah Medical School, IMRIC, Jerusalem, Israel; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Rivka Cohen
- Laboratory of Membrane and Liposome Research, The Hebrew University-Hadassah Medical School, IMRIC, Jerusalem, Israel
| | - Yechezkel Barenholz
- Laboratory of Membrane and Liposome Research, The Hebrew University-Hadassah Medical School, IMRIC, Jerusalem, Israel.
| |
Collapse
|
50
|
Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev 2016. [DOI: '10.1016/j.addr.2015.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
|