1
|
Chan HW, Chow S, Zhang X, Kwok PCL, Chow SF. Role of Particle Size in Translational Research of Nanomedicines for Successful Drug Delivery: Discrepancies and Inadequacies. J Pharm Sci 2023; 112:2371-2384. [PMID: 37453526 DOI: 10.1016/j.xphs.2023.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/30/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Abstract
Despite significant research progress in substantiating the therapeutic merits of nanomedicines and the emergence of sophisticated nanotechnologies, the translation of this knowledge into new therapeutic modalities has been sluggish, indicating the need for a more comprehensive understanding of how the unique physicochemical properties of nanoparticles affect their clinical applications. Particle size is a critical quality attribute that impacts the bio-fate of nanoparticles, yet precise knowledge of its effect remains elusive with discrepancies among literature reports. This review aims to address this scientific knowledge gap from a drug development perspective by highlighting potential inadequacies during the evaluation of particle size effects. We begin with a discussion on the major issues in particle size characterization along with the corresponding remedies. The influence of confounding factors on biological effects of particle size, including colloidal stability, polydispersity, and in vitro drug release, are addressed for establishing stronger in vitro-in vivo correlation. Particle size design and tailoring approaches for successful nanoparticulate drug delivery beyond parenteral administration are also illustrated. We believe a holistic understanding of the effect of particle size on bio-fate, combined with consistent nanoparticle manufacturing platforms and tailored characterization techniques, would expedite the translation of nanomedicines into clinical practice.
Collapse
Affiliation(s)
- Ho Wan Chan
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China
| | - Stephanie Chow
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China
| | - Xinyue Zhang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China; Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, Hong Kong S.A.R, China
| | - Philip Chi Lip Kwok
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Shing Fung Chow
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China; Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, Hong Kong S.A.R, China.
| |
Collapse
|
2
|
Zhu G, Li Z, Zhang Y, Meng X, Guan M, Hu Z, Yang YG, Liu K, Sun T. Biosafety risk assessment of gold and aluminum nanoparticles in tumor-bearing mice. APL Bioeng 2023; 7:016116. [PMID: 36968454 PMCID: PMC10038691 DOI: 10.1063/5.0144481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
To improve the biosafety of the nanodelivery system, this study developed novel monodisperse spherical aluminum nanoparticles (Al NPs) and evaluated their cytotoxicity in vitro and distribution and biotoxicity in vivo. Compared with gold nanoparticles of the same size, Al NPs not only had low cytotoxicity in vitro but also did not cause accumulation in major organs in vivo after intravenous injections. No significant abnormalities were observed in the serum biochemical indices of mice injected with Al NPs. Additionally, no substantial changes occurred in the histopathology of major organs, and no apparent biological toxicity was measured after consecutive injections of Al NPs. These results indicate that Al NPs have a good biological safety and provide a new method for developing low-toxicity nanomedicine.
Collapse
Affiliation(s)
| | - Zhihan Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | - Kun Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, China
- Authors to whom correspondence should be addressed: and
| | - Tianmeng Sun
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
3
|
Sitia G, Fiordaliso F, Violatto MB, Alarcon JF, Talamini L, Corbelli A, Ferreira LM, Tran NL, Chakraborty I, Salmona M, Parak WJ, Diomede L, Bigini P. Food-Grade Titanium Dioxide Induces Toxicity in the Nematode Caenorhabditis elegans and Acute Hepatic and Pulmonary Responses in Mice. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1669. [PMID: 35630890 PMCID: PMC9147568 DOI: 10.3390/nano12101669] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023]
Abstract
Food-grade titanium dioxide (E171) contains variable percentages of titanium dioxide (TiO2) nanoparticles (NPs), posing concerns for its potential effects on human and animal health. Despite many studies, the actual relationship between the physicochemical properties of E171 NPs and their interaction with biological targets is still far from clear. We evaluated the impact of acute E171 administration on invertebrate and vertebrate animals. In the nematode, Caenorhabditis elegans, the administration of up to 1.0 mg/mL of E171 did not affect the worm's viability and lifespan, but significantly impaired its pharyngeal function, reproduction, and development. We also investigated whether the intravenous administration of E171 in mice (at the dose of 6 mg/kg/body weight) could result in an acute over-absorption of filter organs. A significant increase of hepatic titanium concentration and the formation of microgranulomas were observed. Interstitial inflammation and parenchymal modification were found in the lungs, coupled with titanium accumulation. This was probably due to the propensity of TiO2 NPs to agglomerate, as demonstrated by transmission electron microscopy experiments showing that the incubation of E171 with serum promoted the formation of compact clusters. Overall, these data emphasize the actual risk for human and animal exposure to E171.
Collapse
Affiliation(s)
- Giovanni Sitia
- Experimental Hepatology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milano, Italy; (G.S.); (L.M.F.); (N.L.T.)
| | - Fabio Fiordaliso
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (F.F.); (M.B.V.); (J.F.A.); (L.T.); (A.C.); (M.S.)
| | - Martina B. Violatto
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (F.F.); (M.B.V.); (J.F.A.); (L.T.); (A.C.); (M.S.)
| | - Jennifer Fernandez Alarcon
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (F.F.); (M.B.V.); (J.F.A.); (L.T.); (A.C.); (M.S.)
| | - Laura Talamini
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (F.F.); (M.B.V.); (J.F.A.); (L.T.); (A.C.); (M.S.)
| | - Alessandro Corbelli
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (F.F.); (M.B.V.); (J.F.A.); (L.T.); (A.C.); (M.S.)
| | - Lorena Maria Ferreira
- Experimental Hepatology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milano, Italy; (G.S.); (L.M.F.); (N.L.T.)
| | - Ngoc Lan Tran
- Experimental Hepatology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milano, Italy; (G.S.); (L.M.F.); (N.L.T.)
| | - Indranath Chakraborty
- Center for Hybrid Nanostructures (CHyN), Hamburg University, Luruper Chaussee 149, 22607 Hamburg, Germany; (I.C.); (W.J.P.)
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (F.F.); (M.B.V.); (J.F.A.); (L.T.); (A.C.); (M.S.)
| | - Wolfgang J. Parak
- Center for Hybrid Nanostructures (CHyN), Hamburg University, Luruper Chaussee 149, 22607 Hamburg, Germany; (I.C.); (W.J.P.)
| | - Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (F.F.); (M.B.V.); (J.F.A.); (L.T.); (A.C.); (M.S.)
| | - Paolo Bigini
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (F.F.); (M.B.V.); (J.F.A.); (L.T.); (A.C.); (M.S.)
| |
Collapse
|
4
|
Casey LM, Hughes KR, Saunders MN, Miller SD, Pearson RM, Shea LD. Mechanistic contributions of Kupffer cells and liver sinusoidal endothelial cells in nanoparticle-induced antigen-specific immune tolerance. Biomaterials 2022; 283:121457. [PMID: 35286851 PMCID: PMC11225973 DOI: 10.1016/j.biomaterials.2022.121457] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/10/2022] [Accepted: 03/03/2022] [Indexed: 02/06/2023]
Abstract
The intravenous delivery of disease-relevant antigens (Ag) by polymeric nanoparticles (NP-Ags) has demonstrated Ag-specific immune tolerance in autoimmune and allergic disorders as well as allogeneic transplant rejection. NP-Ags are observed to distribute to the spleen, which has an established role in the induction of immune tolerance. However, studies have shown that the spleen is dispensable for NP-Ag-induced tolerance, suggesting significant contributions from other immunological sites. Here, we investigated the tolerogenic contributions of Kupffer cells (KCs) and liver sinusoidal endothelial cells (LSECs) to NP-Ag-induced tolerance in a mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Intravenously delivered Ag-conjugated poly(lactide-co-glycolide) NPs (PLG-Ag) distributed largely to the liver, where they associated with both KCs and LSECs. This distribution was accompanied by CD4 T cell accumulation, clonal deletion, and PD-L1 expression by KCs and LSECs. Ex vivo co-cultures of PLG-Ag-treated KCs or LSECs with Ag-specific CD4 T cells resulted in PGE2 and IL-10 or PGE2 secretion, respectively. KC depletion and adoptive transfer experiments demonstrated that KCs were sufficient, but not necessary, to mediate PLG-Ag-induced tolerance in EAE. The durability of PLG-Ag-induced tolerance in the absence of KCs may be attributed to the distribution of PLG-Ags to LSECs, which demonstrated similar levels of PD-L1, PGE2, and T cell stimulatory ability. Collectively, these studies provide mechanistic support for the role of liver KCs and LSECs in Ag-specific tolerance for a biomaterial platform that is currently being evaluated in clinical trials.
Collapse
Affiliation(s)
- Liam M Casey
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Avenue, Ann Arbor, MI, 48105, USA
| | - Kevin R Hughes
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI, 48109, USA
| | - Michael N Saunders
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI, 48109, USA; Medical Scientist Training Program, University of Michigan, 1135 Catherine St., 2965 Taubman Health Sciences Library, Ann Arbor, MI, 48109, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 6-713 Tarry Building, 303 E. Chicago Avenue, Chicago, IL, 60611, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, 60611, USA
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD, 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA.
| | - Lonnie D Shea
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Avenue, Ann Arbor, MI, 48105, USA; Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
5
|
Karpov TE, Muslimov AR, Antuganov DO, Postovalova AS, Pavlov DA, Usov YV, Shatik SV, Zyuzin MV, Timin AS. Impact of metallic coating on the retention of 225Ac and its daugthers within core-shell nanocarriers. J Colloid Interface Sci 2022; 608:2571-2583. [PMID: 34801240 DOI: 10.1016/j.jcis.2021.10.187] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/14/2021] [Accepted: 10/29/2021] [Indexed: 01/11/2023]
Abstract
Currently, alpha-emitting radionuclide 225Ac is one of the most promising isotopes in alpha therapy due to its high linear energy transfer during four sequential alpha decays. However, the main obstacle preventing the full introduction of 225Ac into clinical practice is the lack of stable retention of radionuclides, leading to free circulation of toxic isotopes in the body. In this work, the surface of silica nanoparticles (SiO2 NPs) has been modified with metallic shells composed of titanium dioxide (TiO2) and gold (Au) nanostructures to improve the retention of 225Ac and its decay products within the developed nanocarriers. In vitro and in vivo studies in healthy mice show that the metallic surface coating of SiO2 NPs promotes an enhanced sequestering of radionuclides (225Ac and its daughter isotopes) compared to non-modified SiO2 NPs for a prolonged period of time. Histological analysis reveals that for the period of 3-10 d after the injections, the developed nanocarriers have no significant toxic effects in mice. At the same time, almost no accumulation of leaked radionuclides can be detected in non-target organs (e.g., in the kidneys). In contrast, non-modified carriers (SiO2 NPs) demonstrate the release of free radionuclides, which are distributed over the whole animal body with the consequent morphological changes in the lung, liver and kidney tissues. These results highlight the potential of the developed nanocarriers to be utilized as radionuclide delivery systems and offer an insight into design rules for the fabrication of new nanotherapeutic agents.
Collapse
Affiliation(s)
- Timofey E Karpov
- Granov Russian Research Center of Radiology & Surgical Technologies, Leningradskaya Street 70 Pesochny, St. Petersburg 197758, Russian Federation; Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg 195251, Russian Federation
| | - Albert R Muslimov
- Granov Russian Research Center of Radiology & Surgical Technologies, Leningradskaya Street 70 Pesochny, St. Petersburg 197758, Russian Federation; St. Petersburg Academic University, Khlopin St. 8/3, St. Petersburg 194021, Russian Federation; Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg 195251, Russian Federation
| | - Dmitrii O Antuganov
- Granov Russian Research Center of Radiology & Surgical Technologies, Leningradskaya Street 70 Pesochny, St. Petersburg 197758, Russian Federation
| | - Alisa S Postovalova
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg 195251, Russian Federation
| | - Dmitri A Pavlov
- Lobachevsky University, 23/3 Gagarin prospect, Nizhny Novgorod 603950, Russian Federation
| | - Yuri V Usov
- Lobachevsky University, 23/3 Gagarin prospect, Nizhny Novgorod 603950, Russian Federation
| | - Sergey V Shatik
- Granov Russian Research Center of Radiology & Surgical Technologies, Leningradskaya Street 70 Pesochny, St. Petersburg 197758, Russian Federation
| | - Mikhail V Zyuzin
- Granov Russian Research Center of Radiology & Surgical Technologies, Leningradskaya Street 70 Pesochny, St. Petersburg 197758, Russian Federation; School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg 191002, Russian Federation.
| | - Alexander S Timin
- Granov Russian Research Center of Radiology & Surgical Technologies, Leningradskaya Street 70 Pesochny, St. Petersburg 197758, Russian Federation; Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg 195251, Russian Federation; Research School of Chemical and Biomedical Engineering, National Research Tomsk Polytechnic University, Lenin Avenue 30, Tomsk 634050, Russian Federation.
| |
Collapse
|
6
|
Shin TH, Manavalan B, Lee DY, Basith S, Seo C, Paik MJ, Kim SW, Seo H, Lee JY, Kim JY, Kim AY, Chung JM, Baik EJ, Kang SH, Choi DK, Kang Y, Maral Mouradian M, Lee G. Silica-coated magnetic-nanoparticle-induced cytotoxicity is reduced in microglia by glutathione and citrate identified using integrated omics. Part Fibre Toxicol 2021; 18:42. [PMID: 34819099 PMCID: PMC8614058 DOI: 10.1186/s12989-021-00433-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/25/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Nanoparticles have been utilized in brain research and therapeutics, including imaging, diagnosis, and drug delivery, owing to their versatile properties compared to bulk materials. However, exposure to nanoparticles leads to their accumulation in the brain, but drug development to counteract this nanotoxicity remains challenging. To date, concerns have risen about the potential toxicity to the brain associated with nanoparticles exposure via penetration of the brain blood barrier to address this issue. METHODS Here the effect of silica-coated-magnetic nanoparticles containing the rhodamine B isothiocyanate dye [MNPs@SiO2(RITC)] were assessed on microglia through toxicological investigation, including biological analysis and integration of transcriptomics, proteomics, and metabolomics. MNPs@SiO2(RITC)-induced biological changes, such as morphology, generation of reactive oxygen species, intracellular accumulation of MNPs@SiO2(RITC) using transmission electron microscopy, and glucose uptake efficiency, were analyzed in BV2 murine microglial cells. Each omics data was collected via RNA-sequencing-based transcriptome analysis, liquid chromatography-tandem mass spectrometry-based proteome analysis, and gas chromatography- tandem mass spectrometry-based metabolome analysis. The three omics datasets were integrated and generated as a single network using a machine learning algorithm. Nineteen compounds were screened and predicted their effects on nanotoxicity within the triple-omics network. RESULTS Intracellular reactive oxygen species production, an inflammatory response, and morphological activation of cells were greater, but glucose uptake was lower in MNPs@SiO2(RITC)-treated BV2 microglia and primary rat microglia in a dose-dependent manner. Expression of 121 genes (from 41,214 identified genes), and levels of 45 proteins (from 5918 identified proteins) and 17 metabolites (from 47 identified metabolites) related to the above phenomena changed in MNPs@SiO2(RITC)-treated microglia. A combination of glutathione and citrate attenuated nanotoxicity induced by MNPs@SiO2(RITC) and ten other nanoparticles in vitro and in the murine brain, protecting mostly the hippocampus and thalamus. CONCLUSIONS Combination of glutathione and citrate can be one of the candidates for nanotoxicity alleviating drug against MNPs@SiO2(RITC) induced detrimental effect, including elevation of intracellular reactive oxygen species level, activation of microglia, and reduction in glucose uptake efficiency. In addition, our findings indicate that an integrated triple omics approach provides useful and sensitive toxicological assessment for nanoparticles and screening of drug for nanotoxicity.
Collapse
Affiliation(s)
- Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Balachandran Manavalan
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Da Yeon Lee
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Shaherin Basith
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Chan Seo
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon, 57922 Republic of Korea
| | - Man Jeong Paik
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon, 57922 Republic of Korea
| | - Sang-Wook Kim
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Haewoon Seo
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Ju Yeon Lee
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanji-ro, Cheongju, 28119 Republic of Korea
| | - Jin Young Kim
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanji-ro, Cheongju, 28119 Republic of Korea
| | - A Young Kim
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Jee Min Chung
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Eun Joo Baik
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Seong Ho Kang
- Department of Chemistry, Graduate School, Kyung Hee University, Yongin-si, Gyeonggi-do 17104 Republic of Korea
- Department of Applied Chemistry and Institute of Natural Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104 Republic of Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, 268 Chungwondaero, Chungju, 27478 Republic of Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - M. Maral Mouradian
- RWJMS Institute for Neurological Therapeutics, Rutgers Biomedical and Health Sciences, and Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854 USA
| | - Gwang Lee
- Department of Molecular Science and Technology, Ajou University, Suwon-si, Gyeonggi-do 16499 Republic of Korea
- Department of Physiology, Ajou University School of Medicine, Suwon-si, Gyeonggi-do 16499 Republic of Korea
| |
Collapse
|
7
|
Chung IJ, Moon H, Jeon SI, Lee HJ, Ahn CH. Ultrasound-triggered imaging and drug delivery using microbubble-self-aggregate complexes. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 33:57-76. [PMID: 34503403 DOI: 10.1080/09205063.2021.1976362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Co-delivery of microbubbles (MBs) with anticancer drugs is a promising theranostic approach that can enhance both the ultrasound contrast and local extravasation of drugs with the sonoporation effect. The simultaneous administration of MBs and hydrophobic drugs, however, is still challenging due to the limitations in drug loading or undesirable stabilization of MBs. In this research, MB-self-aggregate complexes (MB-SAs) were newly fabricated for the encapsulation of hydrophobic drugs, and their theranostic properties are investigated in vitro and in vivo. Glycol chitosan self-aggregates (GC-SAs) loaded with hydrophobic drugs or dyes were chemically conjugated on the surface MBs. Their conjugation ratio was determined to be 73.9%, and GC-SAs on MBs did not affect the stability of MBs. GC-SA attached MBs (GC@MBs) were successfully visualized with low-intensity insonation and showed enhanced cellular uptake via the sonoporation effect. In vivo biodistribution of GC@MBs was examined with tumor-bearing mice, confirming that their accumulation at the tumor site increased by 1.85 times after ultrasound irradiation. The anticancer drug-loaded GC@MBs also exhibited 10% higher cytotoxicity under ultrasound flash. In conclusion, it was expected that GC@MBs could be used both as an ultrasound contrast agent and a drug carrier even with conventional ultrasonic devices.
Collapse
Affiliation(s)
- In Jae Chung
- Department of Materials Science and Engineering, Research Institute of Advanced Materials (RIAM), Seoul National University, Gwanak-gu, Seoul, Korea
| | | | - Seong Ik Jeon
- Department of Materials Science and Engineering, Research Institute of Advanced Materials (RIAM), Seoul National University, Gwanak-gu, Seoul, Korea
| | - Hak Jong Lee
- IMGT Co., Ltd, Seongnam, Korea.,Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Cheol-Hee Ahn
- Department of Materials Science and Engineering, Research Institute of Advanced Materials (RIAM), Seoul National University, Gwanak-gu, Seoul, Korea
| |
Collapse
|
8
|
Dahanayake V, Lyons T, Kerwin B, Rodriguez O, Albanese C, Parasido E, Lee Y, Keuren EV, Li L, Maxey E, Paunesku T, Woloschak G, Stoll SL. Paramagnetic Mn 8Fe 4- co-Polystyrene Nanobeads as a Potential T 1-T 2 Multimodal Magnetic Resonance Imaging Contrast Agent with In Vivo Studies. ACS APPLIED MATERIALS & INTERFACES 2021; 13:39042-39054. [PMID: 34375073 PMCID: PMC10506655 DOI: 10.1021/acsami.1c09232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In developing a cluster-nanocarrier design, as a magnetic resonance imaging contrast agent, we have investigated the enhanced relaxivity of a manganese and iron-oxo cluster grafted within a porous polystyrene nanobead with increased relaxivity due to a higher surface area. The synthesis of the cluster-nanocarrier for the cluster Mn8Fe4O12(O2CC6H4CH═CH2)16(H2O)4, cross-linked with polystyrene (the nanocarrier), under miniemulsion conditions is described. By including a branched hydrophobe, iso-octane, the resulting nanobeads are porous and ∼70 nm in diameter. The increased surface area of the nanobeads compared to nonporous nanobeads leads to an enhancement in relaxivity; r1 increases from 3.8 to 5.2 ± 0.1 mM-1 s-1, and r2 increases from 11.9 to 50.1 ± 4.8 mM-1 s-1, at 9.4 teslas, strengthening the potential for T1 and T2 imaging. Several metrics were used to assess stability, and the porosity produced no reduction in metal stability. Synchrotron X-ray fluorescence microscopy was used to demonstrate that the nanobeads remain intact in vivo. In depth, physicochemical characteristics were determined, including extensive pharmacokinetics, in vivo imaging, and systemic biodistribution analysis.
Collapse
Affiliation(s)
- Vidumin Dahanayake
- Department of Chemistry, Georgetown University, 37th and O Streets NW, Washington, D.C. 20057, United States
| | - Trevor Lyons
- Department of Chemistry, Georgetown University, 37th and O Streets NW, Washington, D.C. 20057, United States
| | - Brendan Kerwin
- Department of Chemistry, Georgetown University, 37th and O Streets NW, Washington, D.C. 20057, United States
| | - Olga Rodriguez
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057, United States
| | - Christopher Albanese
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057, United States
- Department of Radiology, Georgetown University Medical Center, Washington, D.C. 20057, United States
| | - Erika Parasido
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057, United States
| | - Yichien Lee
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057, United States
| | - Edward Van Keuren
- Department of Physics and Institute for Soft Matter Synthesis and Metrology, Georgetown University, 37th and O Streets NW, Washington, D.C. 20057, United States
| | - Luxi Li
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| | - Evan Maxey
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| | - Tatjana Paunesku
- Department of Radiation Oncology, Northwestern University, 303 E. Chicago Ave., Chicago, Illinois 60611, United States
| | - Gayle Woloschak
- Department of Radiation Oncology, Northwestern University, 303 E. Chicago Ave., Chicago, Illinois 60611, United States
| | - Sarah L Stoll
- Department of Chemistry, Georgetown University, 37th and O Streets NW, Washington, D.C. 20057, United States
| |
Collapse
|
9
|
Emerging Nano-Carrier Strategies for Brain Tumor Drug Delivery and Considerations for Clinical Translation. Pharmaceutics 2021; 13:pharmaceutics13081193. [PMID: 34452156 PMCID: PMC8399364 DOI: 10.3390/pharmaceutics13081193] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/13/2022] Open
Abstract
Treatment of brain tumors is challenging since the blood–brain tumor barrier prevents chemotherapy drugs from reaching the tumor site in sufficient concentrations. Nanomedicines have great potential for therapy of brain disorders but are still uncommon in clinical use despite decades of research and development. Here, we provide an update on nano-carrier strategies for improving brain drug delivery for treatment of brain tumors, focusing on liposomes, extracellular vesicles and biomimetic strategies as the most clinically feasible strategies. Finally, we describe the obstacles in translation of these technologies including pre-clinical models, analytical methods and regulatory issues.
Collapse
|
10
|
Tseng YY, Chen TY, Liu SJ. Role of Polymeric Local Drug Delivery in Multimodal Treatment of Malignant Glioma: A Review. Int J Nanomedicine 2021; 16:4597-4614. [PMID: 34267515 PMCID: PMC8275179 DOI: 10.2147/ijn.s309937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022] Open
Abstract
Malignant gliomas (MGs) are the most common and devastating primary brain tumor. At present, surgical interventions, radiotherapy, and chemotherapy are only marginally effective in prolonging the life expectancy of patients with MGs. Inherent heterogeneity, aggressive invasion and infiltration, intact physical barriers, and the numerous mechanisms underlying chemotherapy and radiotherapy resistance contribute to the poor prognosis for patients with MGs. Various studies have investigated methods to overcome these obstacles in MG treatment. In this review, we address difficulties in MG treatment and focus on promising polymeric local drug delivery systems. In contrast to most local delivery systems, which are directly implanted into the residual cavity after intratumoral injection or the surgical removal of a tumor, some rapidly developing and promising nanotechnological methods—including surface-decorated nanoparticles, magnetic nanoparticles, and focused ultrasound assist transport—are administered through (systemic) intravascular injection. We also discuss further synergistic and multimodal strategies for heightening therapeutic efficacy. Finally, we outline the challenges and therapeutic potential of these polymeric drug delivery systems.
Collapse
Affiliation(s)
- Yuan-Yun Tseng
- Department of Neurosurgery, New Taipei Municipal Tu-Cheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei City, Taiwan
| | - Tai-Yuan Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkuo, Tao-Yuan, Taiwan
| |
Collapse
|
11
|
Grun MK, Suberi A, Shin K, Lee T, Gomerdinger V, Moscato ZM, Piotrowski-Daspit AS, Saltzman WM. PEGylation of poly(amine-co-ester) polyplexes for tunable gene delivery. Biomaterials 2021; 272:120780. [PMID: 33813260 DOI: 10.1016/j.biomaterials.2021.120780] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 01/17/2023]
Abstract
There is growing interest in PEGylation of cationic polymeric vehicles for gene delivery in order to improve vehicle stability and reduce toxicity, but little is known about the effects of PEG coatings on transfection. We used a polymer from the poly(amine-co-ester) (PACE) family blended with PEG-conjugated PACE at different ratios in order to explore the effects of polyplex PEGylation on the transfection efficiency of plasmid DNA, mRNA, and siRNA in vitro and mRNA in vivo. We discovered that concentrations of PACE-PEG as low as 0.25% by weight improved polyplex stability but also inhibited transfection in vitro. In vivo, the effect of PACE-PEG incorporation on mRNA transfection varied by delivery route; the addition of PACE-PEG improved local delivery to the lung, but PEGylation had little effect on intravenous systemic delivery. By both delivery routes, transfection was inhibited at concentrations higher than 5 wt% PACE-PEG. These results demonstrate that excess PEGylation can be detrimental to vehicle function, and suggest that PEGylation of cationic vehicles must be optimized by PEG content, cargo type, and delivery route.
Collapse
Affiliation(s)
- Molly K Grun
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, 06511, USA
| | - Alexandra Suberi
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Teresa Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Victoria Gomerdinger
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, 06511, USA
| | - Zoe M Moscato
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | | | - W Mark Saltzman
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, 06511, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA; Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, 06511, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
12
|
Adityan S, Tran M, Bhavsar C, Wu SY. Nano-therapeutics for modulating the tumour microenvironment: Design, development, and clinical translation. J Control Release 2020; 327:512-532. [DOI: 10.1016/j.jconrel.2020.08.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022]
|
13
|
A tumour-selective cascade activatable self-detained system for drug delivery and cancer imaging. Nat Commun 2019; 10:4861. [PMID: 31649241 PMCID: PMC6813295 DOI: 10.1038/s41467-019-12848-5] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 09/26/2019] [Indexed: 12/20/2022] Open
Abstract
Achieving the activation of drugs within cellular systems may provide targeted therapies. Here we construct a tumour-selective cascade activatable self-detained system (TCASS) and incorporate imaging probes and therapeutics. We show in different mouse models that the TCASS system accumulates in solid tumours. The molecules show enhanced accumulation in tumour regions via the effect of recognition induced self-assembly. Analysis of the molecular penetration in tumour tissue shows that in vivo self-assembly increases the penetration capability compared to typical soft or hard nanomaterials. Importantly, the in vivo self-assembled molecules exhibit a comparable clearance pathway to that of small molecules, which are excreted from organs of the reticuloendothelial system (liver and kidney), while are relatively slowly eliminated from tumour tissues. Finally, this system, combined with the NIR probe, shows high specificity and sensitivity for detecting bladder cancer in isolated intact patient bladders. The activation of drugs within cellular systems may provide targeted therapies for cancer. Here, the authors make a drug delivery system that is activated within the cell and exploits XIAP expression to cleave a linker region, resulting in the self-assembly of the system and drug release within cancer cells.
Collapse
|
14
|
Mandl HK, Quijano E, Suh HW, Sparago E, Oeck S, Grun M, Glazer PM, Saltzman WM. Optimizing biodegradable nanoparticle size for tissue-specific delivery. J Control Release 2019; 314:92-101. [PMID: 31654688 DOI: 10.1016/j.jconrel.2019.09.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/03/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023]
Abstract
Nanoparticles (NPs) are promising vehicles for drug delivery because of their potential to target specific tissues [1]. Although it is known that NP size plays a critical role in determining their biological activity, there are few quantitative studies of the role of NP size in determining biodistribution after systemic administration. Here, we engineered fluorescent, biodegradable poly(lactic-co-glycolic acid) (PLGA) NPs in a range of sizes (120-440nm) utilizing a microfluidic platform and used these NPs to determine the effect of diameter on bulk tissue and cellular distribution after systemic administration. We demonstrate that small NPs (∼120nm) exhibit enhanced uptake in bulk lung and bone marrow, while larger NPs are sequestered in the liver and spleen. We also show that small NPs (∼120nm) access specific alveolar cell populations and hematopoietic stem and progenitor cells more readily than larger NPs. Our results suggest that size of PLGA NPs can be used to tune delivery to certain tissues and cell populations in vivo.
Collapse
Affiliation(s)
- Hanna K Mandl
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Elias Quijano
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA; Department of Therapeutic Radiology, Yale University, New Haven, CT, 06520, USA; Department of Genetics, Yale University, New Haven, CT, 06520, USA
| | - Hee Won Suh
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Emily Sparago
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Sebastian Oeck
- Department of Therapeutic Radiology, Yale University, New Haven, CT, 06520, USA
| | - Molly Grun
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Peter M Glazer
- Department of Therapeutic Radiology, Yale University, New Haven, CT, 06520, USA; Department of Genetics, Yale University, New Haven, CT, 06520, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA; Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, 06511, USA; Department of Physiology, Yale University, New Haven, CT, 06511, USA.
| |
Collapse
|
15
|
Prajnamitra RP, Chen HC, Lin CJ, Chen LL, Hsieh PCH. Nanotechnology Approaches in Tackling Cardiovascular Diseases. Molecules 2019; 24:molecules24102017. [PMID: 31137787 PMCID: PMC6572019 DOI: 10.3390/molecules24102017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular diseases have continued to remain a leading cause of mortality and morbidity worldwide. Poor proliferation capability of adult cardiomyocytes disables the heart from regenerating new myocardium after a myocardial ischaemia event and therefore weakens the heart in the long term, which may result in heart failure and death. Delivery of cardioprotective therapeutics soon after the event can help to protect the heart from further cell death and improve cardiac function, but delivery methods and potential side effects of these therapeutics may be an issue. Advances in nanotechnology, particularly nanoparticles for drug delivery, have enabled researchers to obtain better drug targeting capability, thus increasing the therapeutic outcome. Detailed study of nanoparticles in vivo is useful as it can provide insight for future treatments. Nanogel can help to create a more favourable environment, not only for a sustained delivery of therapeutics, but also for a better navigation of the therapeutics to the targeted sites. Finally, if the damage to the myocardium is too severe for drug treatment, nanopatch can help to improve cardiac function and healing by becoming a platform for pluripotent stem cell-derived cardiomyocytes to grow for the purpose of cell-based regenerative therapy.
Collapse
Affiliation(s)
- Ray Putra Prajnamitra
- Institute of Biomedical Sciences, Academia Sinica, 128 Section 2 Academia Road, Nangang District, Taipei 115, Taiwan.
| | - Hung-Chih Chen
- Institute of Biomedical Sciences, Academia Sinica, 128 Section 2 Academia Road, Nangang District, Taipei 115, Taiwan.
| | - Chen-Ju Lin
- Institute of Biomedical Sciences, Academia Sinica, 128 Section 2 Academia Road, Nangang District, Taipei 115, Taiwan.
| | - Li-Lun Chen
- Institute of Biomedical Sciences, Academia Sinica, 128 Section 2 Academia Road, Nangang District, Taipei 115, Taiwan.
| | - Patrick Ching-Ho Hsieh
- Institute of Biomedical Sciences, Academia Sinica, 128 Section 2 Academia Road, Nangang District, Taipei 115, Taiwan.
| |
Collapse
|
16
|
Francini N, Cochrane D, Illingworth S, Purdie L, Mantovani G, Fisher K, Seymour LW, Spain SG, Alexander C. Polyvalent Diazonium Polymers Provide Efficient Protection of Oncolytic Adenovirus Enadenotucirev from Neutralizing Antibodies while Maintaining Biological Activity In Vitro and In Vivo. Bioconjug Chem 2019; 30:1244-1257. [PMID: 30874432 DOI: 10.1021/acs.bioconjchem.9b00189] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Oncolytic viruses offer many advantages for cancer therapy when administered directly to confined solid tumors. However, the systemic delivery of these viruses is problematic because of the host immune response, undesired interactions with blood components, and inherent targeting to the liver. Efficacy of systemically administered viruses has been improved by masking viral surface proteins with polymeric materials resulting in modulation of viral pharmacokinetic profile and accumulation in tumors in vivo. Here we describe a new class of polyvalent reactive polymer based on poly( N-(2-hydroxypropyl)methacrylamide) (polyHPMA) with diazonium reactive groups and their application in the modification of the chimeric group B oncolytic virus enadenotucirev (EnAd). A series of six copolymers with different chain lengths and density of reactive groups was synthesized and used to coat EnAd. Polymer coating was found to be extremely efficient with concentrations as low as 1 mg/mL resulting in complete (>99%) ablation of neutralizing antibody binding. Coating efficiency was found to be dependent on both chain length and reactive group density. Coated viruses were found to have reduced transfection activity both in vitro and in vivo, with greater protection against neutralizing antibodies resulting in lower transgene production. However, in the presence of neutralizing antibodies, some in vivo transgene expression was maintained for coated virus compared to the uncoated control. The decrease in transgene expression was found not to be solely due to lower cellular uptake but due to reduced unpackaging of the virus within the cells and reduced replication, indicating that the polymer coating does not cause permanent inactivation of the virus. These data suggest that virus activity may be modulated by the appropriate design of coating polymers while retaining protection against neutralizing antibodies.
Collapse
Affiliation(s)
- Nora Francini
- School of Pharmacy , University of Nottingham , Nottingham NG7 2RD , U.K
| | - Daniel Cochrane
- PsiOxus Therapeutics Limited , 4-10, The Quadrant, Abingdon Science Park , Abingdon , Oxfordshire OX14 3YS , U.K
| | - Sam Illingworth
- PsiOxus Therapeutics Limited , 4-10, The Quadrant, Abingdon Science Park , Abingdon , Oxfordshire OX14 3YS , U.K
| | - Laura Purdie
- School of Pharmacy , University of Nottingham , Nottingham NG7 2RD , U.K
| | - Giuseppe Mantovani
- School of Pharmacy , University of Nottingham , Nottingham NG7 2RD , U.K
| | - Kerry Fisher
- PsiOxus Therapeutics Limited , 4-10, The Quadrant, Abingdon Science Park , Abingdon , Oxfordshire OX14 3YS , U.K
- Department of Oncology , Old Road Campus Research Building , Roosevelt Drive , Oxford OX3 7DQ , U.K
| | - Leonard W Seymour
- Department of Oncology , Old Road Campus Research Building , Roosevelt Drive , Oxford OX3 7DQ , U.K
| | - Sebastian G Spain
- Department of Chemistry , University of Sheffield , Sheffield S3 7HF , U.K
| | - Cameron Alexander
- School of Pharmacy , University of Nottingham , Nottingham NG7 2RD , U.K
| |
Collapse
|
17
|
Lundy DJ, Chen KH, Toh EKW, Hsieh PCH. Distribution of Systemically Administered Nanoparticles Reveals a Size-Dependent Effect Immediately following Cardiac Ischaemia-Reperfusion Injury. Sci Rep 2016; 6:25613. [PMID: 27161857 PMCID: PMC4861966 DOI: 10.1038/srep25613] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/18/2016] [Indexed: 12/21/2022] Open
Abstract
Nanoparticles represent an attractive option for systemic delivery of therapeutic compounds to the heart following myocardial infarction. However, it is well known that physicochemical properties of nanoparticles such as size, shape and surface modifications can vastly alter the distribution and uptake of injected nanoparticles. Therefore, we aimed to provide an examination of the rapid size-dependent uptake of fluorescent PEG-modified polystyrene nanoparticles administered immediately following cardiac ischaemia-reperfusion injury in mice. By assessing the biodistribution of nanoparticles with core diameters between 20 nm and 2 μm 30 minutes after their administration, we conclude that 20-200 nm diameter nanoparticles are optimal for passive targeting of the injured left ventricle.
Collapse
Affiliation(s)
- David J. Lundy
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Kun-Hung Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Elsie K.-W. Toh
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | | |
Collapse
|
18
|
Evensen L, Johansen PL, Koster G, Zhu K, Herfindal L, Speth M, Fenaroli F, Hildahl J, Bagherifam S, Tulotta C, Prasmickaite L, Mælandsmo GM, Snaar-Jagalska E, Griffiths G. Zebrafish as a model system for characterization of nanoparticles against cancer. NANOSCALE 2016; 8:862-77. [PMID: 26648525 DOI: 10.1039/c5nr07289a] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Therapeutic nanoparticles (NPs) have great potential to deliver drugs against human diseases. Encapsulation of drugs in NPs protects them from being metabolized, while they are delivered specifically to a target site, thereby reducing toxicity and other side-effects. However, non-specific tissue accumulation of NPs, for example in macrophages, especially in the spleen and liver is a general problem with many NPs being developed for cancer therapy. To address the problem of non-specific tissue accumulation of NPs we describe the development of the zebrafish embryo as a transparent vertebrate system for characterization of NPs against cancer. We show that injection of human cancer cells results in tumor-like structures, and that subsequently injected fluorescent NPs, either made of polystyrene or liposomes can be imaged in real-time. NP biodistribution and general in vivo properties can be easily monitored in embryos having selective fluorescent labeling of specific tissues. We demonstrate in vitro, by using optical tweezer micromanipulation, microscopy and flow cytometry that polyethylene glycol (PEG) coating of NPs decreases the level of adhesion of NPs to macrophages, and also to cancer cells. In vivo in zebrafish embryos, PEG coating resulted in longer NP circulation times, decreased macrophage uptake, and reduced adhesion to the endothelium. Importantly, liposomes were observed to accumulate passively and selectively in tumor-like structures comprised of human cancer cells. These results show that zebrafish embryo is a powerful system for microscopy-based screening of NPs on the route to preclinical testing.
Collapse
Affiliation(s)
- Lasse Evensen
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Patrick L Johansen
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Gerbrand Koster
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Kaizheng Zhu
- Department of Chemistry, University of Oslo, Sem Sælands vei 26, 0371, Oslo, Norway
| | - Lars Herfindal
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Martin Speth
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Federico Fenaroli
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Jon Hildahl
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Shahla Bagherifam
- Department of Chemistry, University of Oslo, Sem Sælands vei 26, 0371, Oslo, Norway
| | - Claudia Tulotta
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Lina Prasmickaite
- Department of Tumour Biology, Oslo University Hospital Radiumhospital, Oslo, Norway
| | - Gunhild M Mælandsmo
- Department of Tumour Biology, Oslo University Hospital Radiumhospital, Oslo, Norway
| | - Ewa Snaar-Jagalska
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Gareth Griffiths
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| |
Collapse
|
19
|
Chen KH, Lundy DJ, Toh EKW, Chen CH, Shih C, Chen P, Chang HC, Lai JJ, Stayton PS, Hoffman AS, Hsieh PCH. Nanoparticle distribution during systemic inflammation is size-dependent and organ-specific. NANOSCALE 2015; 7:15863-72. [PMID: 26359216 DOI: 10.1039/c5nr03626g] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
This study comprehensively investigates the changing biodistribution of fluorescent-labelled polystyrene latex bead nanoparticles in a mouse model of inflammation. Since inflammation alters systemic circulatory properties, increases vessel permeability and modulates the immune system, we theorised that systemic inflammation would alter nanoparticle distribution within the body. This has implications for prospective nanocarrier-based therapies targeting inflammatory diseases. Low dose lipopolysaccharide (LPS), a bacterial endotoxin, was used to induce an inflammatory response, and 20 nm, 100 nm or 500 nm polystyrene nanoparticles were administered after 16 hours. HPLC analysis was used to accurately quantify nanoparticle retention by each vital organ, and tissue sections revealed the precise locations of nanoparticle deposition within key tissues. During inflammation, nanoparticles of all sizes redistributed, particularly to the marginal zones of the spleen. We found that LPS-induced inflammation induces splenic macrophage polarisation and alters leukocyte uptake of nanoparticles, with size-dependent effects. In addition, spleen vasculature becomes significantly more permeable following LPS treatment. We conclude that systemic inflammation affects nanoparticle distribution by multiple mechanisms, in a size dependent manner.
Collapse
Affiliation(s)
- K-H Chen
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei 115, Taiwan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Chinen AB, Guan CM, Ferrer JR, Barnaby SN, Merkel TJ, Mirkin CA. Nanoparticle Probes for the Detection of Cancer Biomarkers, Cells, and Tissues by Fluorescence. Chem Rev 2015; 115:10530-74. [PMID: 26313138 DOI: 10.1021/acs.chemrev.5b00321] [Citation(s) in RCA: 629] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Alyssa B Chinen
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chenxia M Guan
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Jennifer R Ferrer
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Stacey N Barnaby
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Timothy J Merkel
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A Mirkin
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
21
|
Colombo L, Zoia L, Violatto MB, Previdi S, Talamini L, Sitia L, Nicotra F, Orlandi M, Salmona M, Recordati C, Bigini P, La Ferla B. Organ Distribution and Bone Tropism of Cellulose Nanocrystals in Living Mice. Biomacromolecules 2015. [DOI: 10.1021/acs.biomac.5b00805] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Laura Colombo
- IRCCS-Istituto
di Ricerche Farmacologiche “Mario Negri”, 20156 Milan, Italy
| | - Luca Zoia
- Department
of Earth and Environmental Science, University of Milano-Bicocca, Piazza
della Scienza 1, 20126 Milan, Italy
| | | | - Sara Previdi
- IRCCS-Istituto
di Ricerche Farmacologiche “Mario Negri”, 20156 Milan, Italy
| | - Laura Talamini
- IRCCS-Istituto
di Ricerche Farmacologiche “Mario Negri”, 20156 Milan, Italy
| | - Leopoldo Sitia
- IRCCS-Istituto
di Ricerche Farmacologiche “Mario Negri”, 20156 Milan, Italy
| | - Francesco Nicotra
- Department
of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza
della Scienza 2, 20126 Milano, Italy
| | - Marco Orlandi
- Department
of Earth and Environmental Science, University of Milano-Bicocca, Piazza
della Scienza 1, 20126 Milan, Italy
| | - Mario Salmona
- IRCCS-Istituto
di Ricerche Farmacologiche “Mario Negri”, 20156 Milan, Italy
| | - Camilla Recordati
- Mouse
and Animal Pathology Laboratory, Fondazione Filarete, Viale Ortles
22/4, 20139 Milano, Italy
| | - Paolo Bigini
- IRCCS-Istituto
di Ricerche Farmacologiche “Mario Negri”, 20156 Milan, Italy
| | - Barbara La Ferla
- Department
of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza
della Scienza 2, 20126 Milano, Italy
| |
Collapse
|
22
|
Combination-targeting to multiple endothelial cell adhesion molecules modulates binding, endocytosis, and in vivo biodistribution of drug nanocarriers and their therapeutic cargoes. J Control Release 2014; 188:87-98. [PMID: 24933603 DOI: 10.1016/j.jconrel.2014.06.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 05/24/2014] [Accepted: 06/07/2014] [Indexed: 01/11/2023]
Abstract
Designing of drug nanocarriers to aid delivery of therapeutics is an expanding field that can improve medical treatments. Nanocarriers are often functionalized with elements that recognize cell-surface molecules involved in subcellular transport to improve targeting and endocytosis of therapeutics. Combination-targeting using several affinity elements further modulates this outcome. The most studied example is endothelial targeting via multiple cell adhesion molecules (CAMs), which mimics the strategy of leukocytes to adhere and traverse the vascular endothelium. Yet, the implications of this strategy on intracellular transport and in vivo biodistribution remain uncharacterized. We examined this using nanocarriers functionalized for dual- or triple-targeting to intercellular, platelet-endothelial, and/or vascular CAMs (ICAM-1, PECAM-1, VCAM-1). These molecules differ in expression level, location, pathological stimulation, and/or endocytic pathway. In endothelial cells, binding of PECAM-1/VCAM-1-targeted nanocarriers was intermediate to single-targeted counterparts and enhanced in disease-like conditions. ICAM-1/PECAM-1-targeted nanocarriers surpassed PECAM-1/VCAM-1 in control, but showed lower selectivity toward disease-like conditions. Triple-targeting resulted in binding similar to ICAM-1/PECAM-1 combination and displayed the highest selectivity in disease-like conditions. All combinations were effectively internalized by the cells, with slightly better performance when targeting receptors of different endocytic pathways. In vivo, ICAM-1/PECAM-1-targeted nanocarriers outperformed PECAM-1/VCAM-1 in control and disease-like conditions, and triple-targeted counterparts slightly enhanced this outcome in some organs. As a result, delivery of a model therapeutic cargo (acid sphingomyelinase, deficient in Niemann-Pick disease A-B) was enhanced to all affected organs by triple-targeted nanocarriers, particularly in disease-like conditions. Therefore, multi-CAM targeting may aid the optimization of some therapeutic nanocarriers, where the combination and multiplicity of the affinity moieties utilized allow modulation of targeting performance.
Collapse
|