1
|
Placci M, Giannotti MI, Muro S. Polymer-based drug delivery systems under investigation for enzyme replacement and other therapies of lysosomal storage disorders. Adv Drug Deliv Rev 2023; 197:114683. [PMID: 36657645 PMCID: PMC10629597 DOI: 10.1016/j.addr.2022.114683] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/30/2022] [Accepted: 12/25/2022] [Indexed: 01/18/2023]
Abstract
Lysosomes play a central role in cellular homeostasis and alterations in this compartment associate with many diseases. The most studied example is that of lysosomal storage disorders (LSDs), a group of 60 + maladies due to genetic mutations affecting lysosomal components, mostly enzymes. This leads to aberrant intracellular storage of macromolecules, altering normal cell function and causing multiorgan syndromes, often fatal within the first years of life. Several treatment modalities are available for a dozen LSDs, mostly consisting of enzyme replacement therapy (ERT) strategies. Yet, poor biodistribution to main targets such as the central nervous system, musculoskeletal tissue, and others, as well as generation of blocking antibodies and adverse effects hinder effective LSD treatment. Drug delivery systems are being studied to surmount these obstacles, including polymeric constructs and nanoparticles that constitute the focus of this article. We provide an overview of the formulations being tested, the diseases they aim to treat, and the results observed from respective in vitro and in vivo studies. We also discuss the advantages and disadvantages of these strategies, the remaining gaps of knowledge regarding their performance, and important items to consider for their clinical translation. Overall, polymeric nanoconstructs hold considerable promise to advance treatment for LSDs.
Collapse
Affiliation(s)
- Marina Placci
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Marina I Giannotti
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; CIBER-BBN, ISCIII, Barcelona, Spain; Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona 08028, Spain
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; Institute of Catalonia for Research and Advanced Studies (ICREA), Barcelona 08010, Spain; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
2
|
Zeng J, Shirihai OS, Grinstaff MW. Modulating lysosomal pH: a molecular and nanoscale materials design perspective. JOURNAL OF LIFE SCIENCES (WESTLAKE VILLAGE, CALIF.) 2020; 2:25-37. [PMID: 33403369 PMCID: PMC7781074 DOI: 10.36069/jols/20201204] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Lysosomes, membrane-bound organelles, play important roles in cellular processes including endocytosis, phagocytosis, and autophagy. Lysosomes maintain cellular homeostasis by generating a highly acidic environment of pH 4.5 - 5.0 and by housing hydrolytic enzymes that degrade engulfed biomolecules. Impairment of lysosomal function, especially in its acidification, is a driving force in the pathogenesis of diseases including neurodegeneration, cancer, metabolic disorders, and infectious diseases. Therefore, lysosomal pH is an attractive and targetable site for therapeutic intervention. Currently, there is a dearth of strategies or materials available to specifically modulate lysosomal acidification. This review focuses on the key aspects of how lysosomal pH is implicated in various diseases and discusses design strategies and molecular or nanoscale agents for lysosomal pH modulation, with the ultimate goal of developing novel therapeutic solutions.
Collapse
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
- Department of Neurology, School of Medicine, Yale University, New Haven, CT 06511
| | - Orian S Shirihai
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90045
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118
- Department of Chemistry, Boston University, Boston, MA 02215
| |
Collapse
|
3
|
Abstract
Liposomes are one of the most widely investigated carriers for CRISPR/Cas9 delivery. The surface properties of liposomal carriers, including the surface charge, PEGylation, and ligand modification can significantly affect the gene silencing efficiency. Three barriers of systemic CRISPR/Cas9 delivery (long blood circulation, efficient tumor penetration, and efficient cellular uptake/endosomal escape) are analyzed on liposomal carriers with different surface charges, PEGylations, and ligand modifications. Cationic formulations dominate CRISPR/Cas9 delivery and neutral formulations also have good performance while anionic formulations are generally not proper for CRISPR/Cas9 delivery. The PEG dilemma (prolonged blood circulation vs. reduced cellular uptake/endosomal escape) and the side effect of repeated PEGylated formulation (accelerated blood clearance) were discussed. Effects of ligand modification on cationic and neutral formulations were analyzed. Finally, we summarized the achievements in liposomal CRISPR/Cas9 delivery, outlined existing problems, and provided some future perspectives. Liposomes are one of the most widely investigated carriers for CRISPR/Cas9 delivery. The surface properties of liposomal carriers, including the surface charge, PEGylation, and ligand modification can significantly affect the gene silencing efficiency. Three barriers of systemic siRNA delivery (long blood circulation, efficient tumor penetration, and efficient cellular uptake/endosomal escape) are analyzed on liposomal carriers with different surface charges, PEGylations, and ligand modifications. Cationic formulations dominate CRISPR/Cas9 delivery and neutral formulations also have good performance while anionic formulations are generally not proper for CRISPR/Cas9 delivery. The PEG dilemma (prolonged blood circulation vs. reduced cellular uptake/endosomal escape) and the side effect of repeated PEGylated formulation (accelerated blood clearance) were discussed. Effects of ligand modification on cationic and neutral formulations were analyzed. Finally, we summarized the achievements in liposomal CRISPR/Cas9 delivery, outlined existing problems, and provided some future perspectives.
Collapse
|
4
|
Gigliobianco MR, Di Martino P, Deng S, Casadidio C, Censi R. New Advanced Strategies for the Treatment of Lysosomal Diseases Affecting the Central Nervous System. Curr Pharm Des 2019; 25:1933-1950. [DOI: 10.2174/1381612825666190708213159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/19/2019] [Indexed: 11/22/2022]
Abstract
Lysosomal Storage Disorders (LSDs), also known as lysosomal diseases (LDs) are a group of serious genetic diseases characterized by not only the accumulation of non-catabolized compounds in the lysosomes due to the deficiency of specific enzymes which usually eliminate these compounds, but also by trafficking, calcium changes and acidification. LDs mainly affect the central nervous system (CNS), which is difficult to reach for drugs and biological molecules due to the presence of the blood-brain barrier (BBB). While some therapies have proven highly effective in treating peripheral disorders in LD patients, they fail to overcome the BBB. Researchers have developed many strategies to circumvent this problem, for example, by creating carriers for enzyme delivery, which improve the enzyme’s half-life and the overexpression of receptors and transporters in the luminal or abluminal membranes of the BBB. This review aims to successfully examine the strategies developed during the last decade for the treatment of LDs, which mainly affect the CNS. Among the LD treatments, enzyme-replacement therapy (ERT) and gene therapy have proven effective, while nanoparticle, fusion protein, and small molecule-based therapies seem to offer considerable promise to treat the CNS pathology. This work also analyzed the challenges of the study to design new drug delivery systems for the effective treatment of LDs. Polymeric nanoparticles and liposomes are explored from their technological point of view and for the most relevant preclinical studies showing that they are excellent choices to protect active molecules and transport them through the BBB to target specific brain substrates for the treatment of LDs.
Collapse
Affiliation(s)
- Maria R. Gigliobianco
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Piera Di Martino
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Siyuan Deng
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Cristina Casadidio
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Roberta Censi
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| |
Collapse
|
5
|
Tuning the Anti(myco)bacterial Activity of 3-Hydroxy-4-pyridinone Chelators through Fluorophores. Pharmaceuticals (Basel) 2018; 11:ph11040110. [PMID: 30347802 PMCID: PMC6316862 DOI: 10.3390/ph11040110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 02/08/2023] Open
Abstract
Controlling the sources of Fe available to pathogens is one of the possible strategies that can be successfully used by novel antibacterial drugs. We focused our interest on the design of chelators to address Mycobacterium avium infections. Taking into account the molecular structure of mycobacterial siderophores and considering that new chelators must be able to compete for Fe(III), we selected ligands of the 3-hydroxy-4-pyridinone class to achieve our purpose. After choosing the type of chelating unit it was also our objective to design chelators that could be monitored inside the cell and for that reason we designed chelators that could be functionalized with fluorophores. We didn’t realize at the time that the incorporation a fluorophore, to allow spectroscopic detection, would be so relevant for the antimycobacterial effect or to determine the affinity of the chelators towards biological membranes. From a biophysical perspective, this is a fascinating illustration of the fact that functionalization of a molecule with a particular label may lead to a change in its membrane permeation properties and result in a dramatic change in biological activity. For that reason we believe it is interesting to give a critical account of our entire work in this area and justify the statement “to label means to change”. New perspectives regarding combined therapeutic approaches and the use of rhodamine B conjugates to target closely related problems such as bacterial resistance and biofilm production are also discussed.
Collapse
|
6
|
Solomon M, Muro S. Lysosomal enzyme replacement therapies: Historical development, clinical outcomes, and future perspectives. Adv Drug Deliv Rev 2017; 118:109-134. [PMID: 28502768 PMCID: PMC5828774 DOI: 10.1016/j.addr.2017.05.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 01/06/2023]
Abstract
Lysosomes and lysosomal enzymes play a central role in numerous cellular processes, including cellular nutrition, recycling, signaling, defense, and cell death. Genetic deficiencies of lysosomal components, most commonly enzymes, are known as "lysosomal storage disorders" or "lysosomal diseases" (LDs) and lead to lysosomal dysfunction. LDs broadly affect peripheral organs and the central nervous system (CNS), debilitating patients and frequently causing fatality. Among other approaches, enzyme replacement therapy (ERT) has advanced to the clinic and represents a beneficial strategy for 8 out of the 50-60 known LDs. However, despite its value, current ERT suffers from several shortcomings, including various side effects, development of "resistance", and suboptimal delivery throughout the body, particularly to the CNS, lowering the therapeutic outcome and precluding the use of this strategy for a majority of LDs. This review offers an overview of the biomedical causes of LDs, their socio-medical relevance, treatment modalities and caveats, experimental alternatives, and future treatment perspectives.
Collapse
Affiliation(s)
- Melani Solomon
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University Maryland, College Park, MD 20742, USA.
| |
Collapse
|
7
|
Costa D, Costa C, Caldeira M, Cortes L, Queiroz JA, Cruz C. Targeting of Cellular Organelles by Fluorescent Plasmid DNA Nanoparticles. Biomacromolecules 2017; 18:2928-2936. [DOI: 10.1021/acs.biomac.7b00877] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Diana Costa
- CICS-UBI
− Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Carolina Costa
- CICS-UBI
− Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Margarida Caldeira
- Microscopy
Unit-CNC − Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Luísa Cortes
- Microscopy
Unit-CNC − Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - João A. Queiroz
- CICS-UBI
− Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Carla Cruz
- CICS-UBI
− Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
8
|
Zhu S, Yao S, Wu F, Jiang L, Wong KL, Zhou J, Wang K. Platinated porphyrin as a new organelle and nucleus dual-targeted photosensitizer for photodynamic therapy. Org Biomol Chem 2017; 15:5764-5771. [PMID: 28660264 DOI: 10.1039/c7ob01003f] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Organelle and nucleus dual-targeted anticancer drugs are being increasingly used for efficient cancer therapy as they can attack the double vital sites of tumor cells. In this work, we synthesized and characterized two new porphyrin compounds Pt-Por-RB and Me-Por-RB. The spectral titration results suggest that both Pt-Por-RB and Me-Por-RB bind to DNA efficiently in an intercalation binding mode. Upon irradiation, Pt-Por-RB with low dark-cytotoxicity can rapidly generate singlet oxygen to damage the tumor cells through the process of photodynamic therapy. Compared with Me-Por-RB, Pt-Por-RB was not only internalized in the organelles, but also in the nuclei of HeLa cells, probably due to the presence of platinum complexes, as analyzed using the confocal laser scanning microscope. Thus, with the combination of organelle and nucleus dual-targeting property and high efficiency of singlet oxygen generation, Pt-Por-RB showed a significant therapeutic activity against tumor cells.
Collapse
Affiliation(s)
- Sizhe Zhu
- Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan, P. R. China.
| | - Si Yao
- Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan, P. R. China.
| | - Fengshou Wu
- Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan, P. R. China.
| | - Lijun Jiang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, P. R. China.
| | - Ka-Leung Wong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, P. R. China.
| | - Ji Zhou
- Hubei Key Lab of Novel Reactor & Green Chemical Technology, Wuhan Institute of Technology, Wuhan, P. R. China
| | - Kai Wang
- Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan, P. R. China. and Hubei Key Lab of Novel Reactor & Green Chemical Technology, Wuhan Institute of Technology, Wuhan, P. R. China
| |
Collapse
|
9
|
Abstract
It is being increasingly recognized that therapeutics need to be delivered to specific organelle targets within cells. Liposomes are versatile lipid-based drug delivery vehicles that can be surface-modified to deliver the loaded cargo to specific subcellular locations within the cell. Hence, the development of such technology requires a means of measuring the subcellular distribution possibly by utilizing imaging techniques that can visualize and quantitate the extent of this subcellular localization. The apparent increase of resolution along the Z-axis offered by confocal microscopy makes this technique suitable for such studies. In this chapter, we describe the application of confocal laser scanning microscopy (CLSM) to determine the subcellular distribution of fluorescently labeled mitochondriotropic liposomes.
Collapse
Affiliation(s)
- Melani A Solomon
- Institute for Bioscience and Biotechnology Research, University of Maryland, 5130 Plant Sciences Building, College Park, MD, 20742, USA.
| |
Collapse
|
10
|
Villamil Giraldo AM, Fyrner T, Wennmalm S, Parikh AN, Öllinger K, Ederth T. Spontaneous Vesiculation and pH-Induced Disassembly of a Lysosomotropic Detergent: Impacts on Lysosomotropism and Lysosomal Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:13566-13575. [PMID: 27936755 DOI: 10.1021/acs.langmuir.6b03458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Lysosomotropic detergents (LDs) selectively rupture lysosomal membranes through mechanisms that have yet to be characterized. A consensus view, currently, holds that LDs, which are weakly basic, diffuse across cellular membranes as monomers in an uncharged state, and via protonation in the acidic lysosomal compartment, they become trapped, accumulate, and subsequently solubilize the membrane and induce lysosomal membrane permeabilization. Here we demonstrate that the lysosomotropic detergent O-methyl-serine dodecylamide hydrochloride (MSDH) spontaneously assembles into vesicles at, and above, cytosolic pH, and that the vesicles disassemble as the pH reaches 6.4 or lower. The aggregation commences at concentrations below the range of those used in cell studies. Assembly and disassembly of the vesicles was studied via dynamic light scattering, zeta potential measurements, cryo-TEM, and fluorescence correlation spectroscopy and was found to be reversible via control of the pH. Aggregation of MSDH into closed vesicles under cytosolic conditions is at variance with the commonly held view of LD behavior, and we propose that endocytotic pathways should be considered as possible routes of LD entry into lysosomes. We further demonstrate that MSDH vesicles can be loaded with fluorophores via a solution transition from low to high pH, for subsequent release when the pH is lowered again. The ability to encapsulate molecular cargo into MSDH vesicles together with its ability to disaggregate at low pH and to permeabilize the lysosomal membrane presents an intriguing possibility to use MSDH as a delivery system.
Collapse
Affiliation(s)
- Ana M Villamil Giraldo
- Experimental Pathology, Department of Clinical and Experimental Medicine, Linköping, University , SE-581 85 Linköping, Sweden
| | | | - Stefan Wennmalm
- Royal Institute of Technology, Department of Applied Physics, Experimental Biomolecular Physics, Scilifelab , 171 65 Solna, Sweden
| | - Atul N Parikh
- Departments of Biomedical Engineering and Materials Science & Engineering, University of California , Davis, California 95616, United States
| | - Karin Öllinger
- Experimental Pathology, Department of Clinical and Experimental Medicine, Linköping, University , SE-581 85 Linköping, Sweden
| | | |
Collapse
|
11
|
Giannotti MI, Abasolo I, Oliva M, Andrade F, García-Aranda N, Melgarejo M, Pulido D, Corchero JL, Fernández Y, Villaverde A, Royo M, García-Parajo MF, Sanz F, Schwartz S. Highly Versatile Polyelectrolyte Complexes for Improving the Enzyme Replacement Therapy of Lysosomal Storage Disorders. ACS APPLIED MATERIALS & INTERFACES 2016; 8:25741-25752. [PMID: 27610822 DOI: 10.1021/acsami.6b08356] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Lysosomal storage disorders are currently treated by enzyme replacement therapy (ERT) through the direct administration of the unprotected recombinant protein to the patients. Herein we present an ionically cross-linked polyelectrolyte complex (PEC) composed of trimethyl chitosan (TMC) and α-galactosidase A (GLA), the defective enzyme in Fabry disease, with the capability of directly targeting endothelial cells by incorporating peptide ligands containing the RGD sequence. We assessed the physicochemical properties, cytotoxicity, and hemocompatibility of RGD-targeted and untargeted PECs, the uptake by endothelial cells and the intracellular activity of PECs in cell culture models of Fabry disease. Moreover, we also explored the effect of different freeze-drying procedures in the overall activity of the PECs. Our results indicate that the use of integrin-binding RGD moiety within the PEC increases their uptake and the efficacy of the GLA enzyme, while the freeze-drying allows the activity of the therapeutic protein to remain intact. Overall, these results highlight the potential of TMC-based PECs as a highly versatile and feasible drug delivery system for improving the ERT of lysosomal storage disorders.
Collapse
Affiliation(s)
- Marina I Giannotti
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Physical Chemistry Department, Universitat de Barcelona , 08028 Barcelona, Spain
| | - Ibane Abasolo
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| | - Mireia Oliva
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Pharmacy and Pharmaceutical Technology Department, Universitat de Barcelona , 08028 Barcelona, Spain
| | - Fernanda Andrade
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Pharmacy and Pharmaceutical Technology Department, Universitat de Barcelona , 08028 Barcelona, Spain
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto , 4050-313 Porto, Portugal
| | - Natalia García-Aranda
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| | - Marta Melgarejo
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Combinatorial Chemistry Unit, Barcelona Science Park , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Daniel Pulido
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Combinatorial Chemistry Unit, Barcelona Science Park , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - José L Corchero
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona , Bellaterra, 08193 Cerdanyola del Vallès, Spain
| | - Yolanda Fernández
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| | - Antonio Villaverde
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona , Bellaterra, 08193 Cerdanyola del Vallès, Spain
| | - Miriam Royo
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Combinatorial Chemistry Unit, Barcelona Science Park , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - María F García-Parajo
- Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology , ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Fausto Sanz
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Physical Chemistry Department, Universitat de Barcelona , 08028 Barcelona, Spain
| | - Simó Schwartz
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| |
Collapse
|
12
|
Cordova A, Woodrick J, Grindrod S, Zhang L, Saygideger-Kont Y, Wang K, DeVito S, Daniele SG, Paige M, Brown ML. Aminopeptidase P Mediated Targeting for Breast Tissue Specific Conjugate Delivery. Bioconjug Chem 2016; 27:1981-90. [PMID: 26965452 DOI: 10.1021/acs.bioconjchem.5b00481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cytotoxic chemotherapies are used to treat breast cancer, but are limited by systemic toxicity. The key to addressing this important issue is the development of a nontoxic, tissue selective, and molecular specific delivery system. In order to potentially increase the therapeutic index of clinical reagents, we designed an Aminopeptidase P (APaseP) targeting tissue-specific construct conjugated to a homing peptide for selective binding to human breast-derived cancer cells. Homing peptides are short amino acid sequences derived from phage display libraries that have the unique property of localizing to specific organs. Our molecular construct allows for tissue-specific drug delivery, by binding to APaseP in the vascular endothelium. The breast homing peptide evaluated in our studies is a cyclic nine-amino-acid peptide with the sequence CPGPEGAGC, referred to as PEGA. We show by confocal microscopy that the PEGA peptide and similar peptide conjugates distribute to human breast tissue xenograft specifically and evaluate the interaction with the membrane-bound proline-specific APaseP (KD = 723 ± 3 nM) by binding studies. To achieve intracellular breast cancer cell delivery, the incorporation of the Tat sequence, a cell-penetrating motif derived from HIV, was conjugated with the fluorescently labeled PEGA peptide sequence. Ultimately, tissue specific peptides and their conjugates can enhance drug delivery and treatment by their ability to discriminate between tissue types. Tissue specific conjugates as we have designed may be valuable tools for drug delivery and visualization, including the potential to treat breast cancer, while simultaneously minimizing systemic toxicity.
Collapse
Affiliation(s)
- Antoinette Cordova
- Center for Drug Discovery, Georgetown University Medical Center , 3970 Reservoir Road NW Washington, DC 20057, United States
| | - Jordan Woodrick
- Georgetown University Medical Center , 3900 Reservoir Road NW, Washington, DC 20057, United States
| | - Scott Grindrod
- Center for Drug Discovery, Georgetown University Medical Center , 3970 Reservoir Road NW Washington, DC 20057, United States
| | - Li Zhang
- Center for Drug Discovery, Georgetown University Medical Center , 3970 Reservoir Road NW Washington, DC 20057, United States
| | - Yasemin Saygideger-Kont
- Georgetown University Medical Center , 3900 Reservoir Road NW, Washington, DC 20057, United States
| | - Kan Wang
- Center for Drug Discovery, Georgetown University Medical Center , 3970 Reservoir Road NW Washington, DC 20057, United States
| | - Stephen DeVito
- Georgetown University Medical Center , 3900 Reservoir Road NW, Washington, DC 20057, United States
| | - Stefano G Daniele
- Georgetown University Medical Center , 3900 Reservoir Road NW, Washington, DC 20057, United States
| | - Mikell Paige
- George Mason University , Department of Chemistry and Biochemistry, 10900 University Boulevard, Manassas, Virginia 20110, United States
| | - Milton L Brown
- Center for Drug Discovery, Georgetown University Medical Center , 3970 Reservoir Road NW Washington, DC 20057, United States
| |
Collapse
|
13
|
Zylberberg C, Matosevic S. Pharmaceutical liposomal drug delivery: a review of new delivery systems and a look at the regulatory landscape. Drug Deliv 2016; 23:3319-3329. [PMID: 27145899 DOI: 10.1080/10717544.2016.1177136] [Citation(s) in RCA: 369] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Liposomes were the first nanoscale drug to be approved for clinical use in 1995. Since then, the technology has grown considerably, and pioneering recent work in liposome-based delivery systems has brought about remarkable developments with significant clinical implications. This includes long-circulating liposomes, stimuli-responsive liposomes, nebulized liposomes, elastic liposomes for topical, oral and transdermal delivery and covalent lipid-drug complexes for improved drug plasma membrane crossing and targeting to specific organelles. While the regulatory bodies' opinion on liposomes is well-documented, current guidance that address new delivery systems are not. This review describes, in depth, the current state-of-the-art of these new liposomal delivery systems and provides a critical overview of the current regulatory landscape surrounding commercialization efforts of higher-level complexity systems, the expected requirements and the hurdles faced by companies seeking to bring novel liposome-based systems for clinical use to market.
Collapse
|
14
|
|
15
|
Qiu K, Liu Y, Huang H, Liu C, Zhu H, Chen Y, Ji L, Chao H. Biscylometalated iridium(iii) complexes target mitochondria or lysosomes by regulating the lipophilicity of the main ligands. Dalton Trans 2016; 45:16144-16147. [DOI: 10.1039/c6dt03328h] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An efficient method that controls biscylometalated iridium(iii) complexes to target mitochondria or lysosomes was presented.
Collapse
Affiliation(s)
- Kangqiang Qiu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Yukang Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Huaiyi Huang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Chaofeng Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Hongyi Zhu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| |
Collapse
|
16
|
Wexselblatt E, Esko JD, Tor Y. GNeosomes: Highly Lysosomotropic Nanoassemblies for Lysosomal Delivery. ACS NANO 2015; 9:3961-3968. [PMID: 25831231 DOI: 10.1021/nn507382n] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
GNeosomes, lysosomotropic lipid vesicles decorated with guanidinoneomycin, can encapsulate and facilitate the cellular internalization and lysosomal delivery of cargo ranging from small molecules to high molecular weight proteins, in a process that is exclusively dependent on cell surface glycosaminoglycans. Their cellular uptake mechanism and co-localization with lysosomes, as well as the delivery, release, and activity of internalized cargo, are quantified. GNeosomes are proposed as a universal platform for lysosomal delivery with potential as a basic research tool and a therapeutic vehicle.
Collapse
Affiliation(s)
- Ezequiel Wexselblatt
- †Chemistry and Biochemistry and ‡Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jeffrey D Esko
- †Chemistry and Biochemistry and ‡Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Yitzhak Tor
- †Chemistry and Biochemistry and ‡Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
17
|
Wang A, Guo M, Wang N, Zhao J, Qi W, Muhammad F, Chen L, Guo Y, Nguyen NT, Zhu G. Redox-mediated dissolution of paramagnetic nanolids to achieve a smart theranostic system. NANOSCALE 2014; 6:5270-5278. [PMID: 24695417 DOI: 10.1039/c3nr05687b] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Manganese oxide (Mn3O4) nanoparticles have recently emerged as a promising T1 contrast agent. In this study, for the first time, we demonstrated an interaction of Mn3O4 with a biological system, and found redox sensitive behavior of these paramagnetic nanoparticles in intracellular reducing environment. Inspired by these findings, we for the first time used this interaction for some therapeutic advantages and designed a versatile mesoporous silica based nanotheranostic system to realize redox-activated enhanced magnetic resonance imaging and responsive anticancer drug delivery. Contrary to previous reports, we firstly prepared high quality amine terminated hydrophilic Mn3O4 nanolids, without using multistep ligand exchange strategies. The resulting water stable and small-sized Mn3O4 nanolids were subsequently used as nanolids to cap drug loaded nanochannels of a porous carrier. Exposure to highly prevalent intracellular reducing environment resulted in the steady-state dissolution of these nanolids and attained an intelligent drug release. Furthermore, the redox receptive dissolution of paramagnetic Mn3O4 nanolids into Mn(2+) in turn increases the T1 signal to twofold, providing an added opportunity to even track the feedback of therapy. This study, in addition to simultaneously realizing drug delivery and imaging, also provides a new insight into the fate and interaction of manganese oxide nanoparticles with components of biological systems.
Collapse
Affiliation(s)
- Aifei Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Lan H, Wen Y, Shi Y, Liu K, Mao Y, Yi T. Fluorescence turn-on detection of Sn2+ in live eukaryotic and prokaryotic cells. Analyst 2014; 139:5223-9. [DOI: 10.1039/c4an01014k] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fluorescence turn-on probes for selective detection of Sn(ii) in live eukaryotic and prokaryotic cells were developed.
Collapse
Affiliation(s)
- Haichuang Lan
- Department of Chemistry and Concerted Innovation Center of Chemistry for Energy Materials
- Fudan University
- Shanghai 200433, China
| | - Ying Wen
- Department of Chemistry and Concerted Innovation Center of Chemistry for Energy Materials
- Fudan University
- Shanghai 200433, China
| | - Yunming Shi
- P&G Technology (Beijing) Co., Ltd
- Beijing, China
| | - Keyin Liu
- Department of Chemistry and Concerted Innovation Center of Chemistry for Energy Materials
- Fudan University
- Shanghai 200433, China
| | - Yueyuan Mao
- Department of Chemistry and Concerted Innovation Center of Chemistry for Energy Materials
- Fudan University
- Shanghai 200433, China
| | - Tao Yi
- Department of Chemistry and Concerted Innovation Center of Chemistry for Energy Materials
- Fudan University
- Shanghai 200433, China
| |
Collapse
|
19
|
Sun D, Liu Y, Yu Q, Qin X, Yang L, Zhou Y, Chen L, Liu J. Inhibition of tumor growth and vasculature and fluorescence imaging using functionalized ruthenium-thiol protected selenium nanoparticles. Biomaterials 2013; 35:1572-83. [PMID: 24268198 DOI: 10.1016/j.biomaterials.2013.11.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 11/02/2013] [Indexed: 12/12/2022]
Abstract
Here we reported the high tumor targeting efficacy of luminescent Ru(II)-thiols protected selenium nanoparticles (Ru-MUA@Se). We have shown that a dual-target inhibitor Ru-MUA@Se directly suppress the tumor growth but also block blood-vessel growth. We also determined that the nanoparticles entered the cells via clathrin-mediated endocytosis pathway. In a xenograft HepG2 tumor model, we found that Ru-MUA@Se effectively inhibited tumor angiogenesis and suppressed tumor growth with low side effects using metronomic chemotherapy with Ru-MUA@Se. In vivo investigation of nanoparticles on nude mice bearing HepG2 cancer xenografts confirmed that Ru-MUA@Se nanoparticles possessed high tumor-targeted fluorescence imaging, exhibited enhanced antitumor efficacy and decreased systemic toxicity. Moreover, Ru-MUA@Se not only significantly induced dose-dependent disruption of mitochondrial membrane potential in HepG2 cells after 24 h treatment, but it also enhanced reactive oxygen species (ROS) generation. Our results suggest that the potential application of these Ru-MUA@Se nanoparticles in targeting cancer imaging and chemotherapy.
Collapse
Affiliation(s)
- Dongdong Sun
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yanan Liu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Qianqian Yu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Xiuying Qin
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Licong Yang
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yanhui Zhou
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Lanmei Chen
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Jie Liu
- Department of Chemistry, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
20
|
Thekkedath R, Koshkaryev A, Torchilin VP. Lysosome-targeted octadecyl-rhodamine B-liposomes enhance lysosomal accumulation of glucocerebrosidase in Gaucher's cells in vitro. Nanomedicine (Lond) 2012. [PMID: 23199221 DOI: 10.2217/nnm.12.138] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
AIM We hypothesized that liposomes modified with lysosomotropic octadecyl-rhodamine B (Rh) and loaded with therapeutic glucocerebroside velaglucerase alfa (VPRIV™) will improve lysosomal delivery of the enzyme into Gaucher's cells. MATERIALS & METHODS Confocal microscopy and flow cytometry were used to evaluate the ability of Rh-modified liposomes loaded with VPRIV to improve the lysosomal targeting in monocyte-derived macrophages and Gaucher's fibroblasts. RESULTS Confocal microscopy demonstrated that Rh-modified liposomes localized primarily in the lysosomes. As confirmed by flow cytometry using specific substrate 5-(pentafluorobenzoylamino)fluorescein diglucoside, intralysosomal accumulation of VPRIV in the cells treated with Rh-modified liposomes was significantly increased (up to 68%) relative to the cells treated with plain liposomes or free VPRIV. CONCLUSION Rh-modified lysosomotropic liposomes can improve lysosomal accumulation of liposomal enzymes both in nonphagocytic Gaucher's fibroblasts and phagocytic monocyte-derived macrophages.
Collapse
Affiliation(s)
- Ritesh Thekkedath
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology & Nanomedicine, Northeastern University, 140 Fenway Street, Boston, MA 02115, USA
| | | | | |
Collapse
|
21
|
|
22
|
Duncan R, Richardson SCW. Endocytosis and intracellular trafficking as gateways for nanomedicine delivery: opportunities and challenges. Mol Pharm 2012; 9:2380-402. [PMID: 22844998 DOI: 10.1021/mp300293n] [Citation(s) in RCA: 248] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
More than 40 nanomedicines are already in routine clinical use with a growing number following in preclinical and clinical development. The therapeutic objectives are often enhanced disease-specific targeting (with simultaneously reduced access to sites of toxicity) and, especially in the case of macromolecular biotech drugs, improving access to intracellular pharmacological target receptors. Successful navigation of the endocytic pathways is usually a prerequisite to achieve these goals. Thus a comprehensive understanding of endocytosis and intracellular trafficking pathways in both the target and bystander normal cell type(s) is essential to enable optimal nanomedicine design. It is becoming evident that endocytic pathways can become disregulated in disease and this, together with the potential changes induced during exposure to the nanocarrier itself, has the potential to significantly impact nanomedicine performance in terms of safety and efficacy. Here we overview the endomembrane trafficking pathways, discuss the methods used to determine and quantitate the intracellular fate of nanomedicines, and review the current status of lysosomotropic and endosomotropic delivery. Based on the lessons learned during more than 3 decades of clinical development, the need to use endocytosis-relevant clinical biomarkers to better select those patients most likely to benefit from nanomedicine therapy is also discussed.
Collapse
Affiliation(s)
- Ruth Duncan
- School of Science, University of Greenwich, Central Avenue, Chatham Maritime, Kent, ME4 4TB, UK.
| | | |
Collapse
|