1
|
Sandvig K, Iversen TG, Skotland T. Entry of nanoparticles into cells and tissues: status and challenges. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:1017-1029. [PMID: 39161463 PMCID: PMC11331539 DOI: 10.3762/bjnano.15.83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024]
Abstract
In this article we discuss how nanoparticles (NPs) of different compositions may interact with and be internalized by cells, and the consequences of that for cellular functions. A large number of NPs are made with the intention to improve cancer treatment, the goal being to increase the fraction of injected drug delivered to the tumor and thereby improve the therapeutic effect and decrease side effects. Thus, we discuss how NPs are delivered to tumors and some challenges related to investigations of biodistribution, pharmacokinetics, and excretion. Finally, we discuss requirements for bringing NPs into clinical use and aspects when it comes to usage of complex and slowly degraded or nondegradable NPs.
Collapse
Affiliation(s)
- Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, 0379 Oslo, Norway
- Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| | - Tore Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, 0379 Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, 0379 Oslo, Norway
| |
Collapse
|
2
|
Snipstad S, Sulheim E, Åslund AKO, Hyldbakk A, Wågbø AM, Klinkenberg G, Mørch Y. Nanoparticle-loaded microbubbles for treatment of lung cancer. Eur J Pharm Sci 2024; 199:106804. [PMID: 38763448 DOI: 10.1016/j.ejps.2024.106804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/20/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Lung cancer is one of the most common cancers and a leading cause of death, with poor prognosis and high unmet clinical need. Chemotherapy is a common part of the treatment, either alone or in combination with other treatment modalities, but with limited efficacy and severe side effects. Encapsulation of drugs into nanoparticles can enable a more targeted delivery with reduced off-target toxicity. Delivery to the lungs is however often insufficient due to various biological barriers in the body and in the tumor microenvironment. Here we demonstrate that by incorporating drug-loaded nanoparticles into air-filled microbubbles, a more effective targeting to the lungs can be achieved. Fluorescence imaging and mass spectrometry revealed that the microbubbles could significantly improve accumulation of drug in the lungs of mice, compared to injecting either the free drug by itself or only the drug-loaded nanoparticles. Therapeutic efficacy was verified in a preclinical mouse model with non-small cell lung cancer, monitoring tumor growth by luminescence.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olavs Hospital, Trondheim, Norway.
| | - Einar Sulheim
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Andreas K O Åslund
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Astrid Hyldbakk
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ane Marit Wågbø
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Geir Klinkenberg
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; NaDeNo Nanoscience AS, Trondheim, Norway
| |
Collapse
|
3
|
Marzi A, Eder KM, Barroso Á, Kemper B, Schnekenburger J. Quantitative Phase Imaging as Sensitive Screening Method for Nanoparticle-Induced Cytotoxicity Assessment. Cells 2024; 13:697. [PMID: 38667312 PMCID: PMC11049110 DOI: 10.3390/cells13080697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The assessment of nanoparticle cytotoxicity is challenging due to the lack of customized and standardized guidelines for nanoparticle testing. Nanoparticles, with their unique properties, can interfere with biochemical test methods, so multiple tests are required to fully assess their cellular effects. For a more reliable and comprehensive assessment, it is therefore imperative to include methods in nanoparticle testing routines that are not affected by particles and allow for the efficient integration of additional molecular techniques into the workflow. Digital holographic microscopy (DHM), an interferometric variant of quantitative phase imaging (QPI), has been demonstrated as a promising method for the label-free assessment of the cytotoxic potential of nanoparticles. Due to minimal interactions with the sample, DHM allows for further downstream analyses. In this study, we investigated the capabilities of DHM in a multimodal approach to assess cytotoxicity by directly comparing DHM-detected effects on the same cell population with two downstream biochemical assays. Therefore, the dry mass increase in RAW 264.7 macrophages and NIH-3T3 fibroblast populations measured by quantitative DHM phase contrast after incubation with poly(alkyl cyanoacrylate) nanoparticles for 24 h was compared to the cytotoxic control digitonin, and cell culture medium control. Viability was then determined using a metabolic activity assay (WST-8). Moreover, to determine cell death, supernatants were analyzed for the release of the enzyme lactate dehydrogenase (LDH assay). In a comparative analysis, in which the average half-maximal effective concentration (EC50) of the nanocarriers on the cells was determined, DHM was more sensitive to the effect of the nanoparticles on the used cell lines compared to the biochemical assays.
Collapse
Affiliation(s)
- Anne Marzi
- Biomedical Technology Center, University of Muenster, Mendelstraße 17, D-48149 Muenster, Germany; (K.M.E.); (Á.B.); (B.K.)
| | | | | | | | | |
Collapse
|
4
|
Valsalakumari R, Pandya AD, Prasmickaite L, Kvalvaag A, Myrann AG, Åslund AKO, Kjos MS, Fontecha-Cuenca C, Haroon HB, Ribeiro ARS, Horejs-Hoeck J, Moghimi SM, Mørch Ý, Skotland T, Sandvig K, Mælandsmo GM, Iversen TG. Preclinical Efficacy of Cabazitaxel Loaded Poly(2-alkyl cyanoacrylate) Nanoparticle Variants. Int J Nanomedicine 2024; 19:3009-3029. [PMID: 38562610 PMCID: PMC10982070 DOI: 10.2147/ijn.s450283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/09/2024] [Indexed: 04/04/2024] Open
Abstract
Background Biodegradable poly(alkyl cyanoacrylate) (PACA) nanoparticles (NPs) are receiving increasing attention in anti-cancer nanomedicine development not only for targeted cancer chemotherapy, but also for modulation of the tumor microenvironment. We previously reported promising results with cabazitaxel (CBZ) loaded poly(2-ethylbutyl cyanoacrylate) NPs (PEBCA-CBZ NPs) in a patient derived xenograft (PDX) model of triple-negative breast cancer, and this was associated with a decrease in M2 macrophages. The present study aims at comparing two endotoxin-free PACA NP variants (PEBCA and poly(2-ethylhexyl cyanoacrylate); PEHCA), loaded with CBZ and test whether conjugation with folate would improve their effect. Methods Cytotoxicity assays and cellular uptake of NPs by flow cytometry were performed in different breast cancer cells. Biodistribution and efficacy studies were performed in PDX models of breast cancer. Tumor associated immune cells were analyzed by multiparametric flow cytometry. Results In vitro studies showed similar NP-induced cytotoxicity patterns despite difference in early NP internalization. On intravenous injection, the liver cleared the majority of NPs. Efficacy studies in the HBCx39 PDX model demonstrated an enhanced effect of drug-loaded PEBCA variants compared with free drug and PEHCA NPs. Furthermore, the folate conjugated PEBCA variant did not show any enhanced effects compared with the unconjugated counterpart which might be due to unfavorable orientation of folate on the NPs. Finally, analyses of the immune cell populations in tumors revealed that treatment with drug loaded PEBCA variants affected the myeloid cells, especially macrophages, contributing to an inflammatory, immune activated tumor microenvironment. Conclusion We report for the first time, comparative efficacy of PEBCA and PEHCA NP variants in triple negative breast cancer models and show that CBZ-loaded PEBCA NPs exhibit a combined effect on tumor cells and on the tumor associated myeloid compartment, which may boost the anti-tumor response.
Collapse
Affiliation(s)
- Remya Valsalakumari
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, Oslo, 0379, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
| | - Abhilash D Pandya
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
| | - Lina Prasmickaite
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
| | - Audun Kvalvaag
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, Oslo, 0379, Norway
| | - Anne Grethe Myrann
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, Oslo, 0379, Norway
| | - Andreas K O Åslund
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, 7034, Norway
| | | | - Cristina Fontecha-Cuenca
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
- Department of Biomedical Science, University of Padova, Padova, Italy
| | - Hajira B Haroon
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Ana R S Ribeiro
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Salzburg, 5020, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - S Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
- Faculty of Health and Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, 7034, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, Oslo, 0379, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, Oslo, 0379, Norway
- Department of Biosciences, University of Oslo, Oslo, 0316, Norway
| | - Gunhild Mari Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
- Department of Medical Biology, University of Tromsø, Tromsø, 9019, Norway
| | - Tore Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
- Centre for Cancer Cell Reprogramming, University of Oslo, Oslo, 0379, Norway
| |
Collapse
|
5
|
Hyldbakk A, Hansen T, Hak S, Borgos SEF. Polyethylene glycol (PEG) as a broad applicability marker for LC-MS/MS-based biodistribution analysis of nanomedicines. J Control Release 2024; 366:611-620. [PMID: 38215988 DOI: 10.1016/j.jconrel.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/15/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
Polyethylene glycol (PEG) conjugation (PEGylation) is a well-established strategy to improve the pharmacokinetic and biocompatibility properties of a wide variety of nanomedicines and therapeutic peptides and proteins. This broad use makes PEG an attractive 'allround' candidate marker for the biodistribution of such PEGylated compounds. This paper presents the development of a novel strategy for PEG quantification in biological matrices. The methodology is based on sample hydrolysis which both decomposes the sample matrix and degrades PEGylated analytes to specific molecular fragments more suitable for detection by LC-MS/MS. Method versatility was demonstrated by applying it to a wide variety of PEGylated compounds, including polymeric poly(ethylbutyl cyanoacrylate) (PEBCA) nanoparticles, lipidic nanoparticles (Doxil®, LipImage 815™ and lipid nanoparticles for nucleic acid delivery) and the antibody Cimzia®. Method applicability was assessed by analyzing plasma and tissue samples from a comprehensive drug biodistribution study in rats, of both PEBCA and LipImage 815™ nanoparticles. The results demonstrated the method's utility for biodistribution studies on PEG. Importantly, by using the method described herein in tandem with quantification of nanoparticle payloads, we showed that this approach can provide detailed understanding of various critical aspects of the in vivo behavior of PEGylated nanomedicines, such as drug release and particle stability. Together, the presented results demonstrate the novel method as a robust, versatile and generic approach for biodistribution analysis of PEGylated therapeutics.
Collapse
Affiliation(s)
- Astrid Hyldbakk
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Terkel Hansen
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Sjoerd Hak
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sven Even F Borgos
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway.
| |
Collapse
|
6
|
Ahiable MG, Matsunaga K, Hokin M, Iida K, Befu F, Oshima SI. In Vitro Efficacy of Isobutyl Cyanoacrylate Nanoparticles against Fish Bacterial Pathogens and Selection Preference by Rainbow Trout ( Oncorhynchus mykiss). Microorganisms 2023; 11:2877. [PMID: 38138020 PMCID: PMC10745873 DOI: 10.3390/microorganisms11122877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 12/24/2023] Open
Abstract
The upsurge in havoc being wreaked by antibiotic-resistant bacteria has led to an urgent need for efficacious alternatives to antibiotics. This study assessed the antibacterial efficacy of two isobutyl cyanoacrylate nanoparticles (iBCA-NPs), D6O and NP30, against major bacterial pathogens of fish. In vivo tests on rainbow trout were preceded by in vitro tests of minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC). NP30 exhibited higher efficacy than D60, but both iBCA-NPs demonstrated dose-dependent and species-specific in vitro antibacterial properties against the bacterial isolates. Generally, Gram-negative bacteria were more resistant to the iBCA-NPs. Streptococcus iniae, Tenacibaculum maritimum, and Photobacterium damselae were particularly sensitive to both iBCA-NPs. Administered to rainbow trout at 3571.4 mg (iBCA-NP)/kg feed, the iBCA-NPs produced a relative gain rate and survival rates comparable to the control (p > 0.05). The condition factor and the hepatosomatic and viscerosomatic indices of fish were indifferentiable (p > 0.05) between the iBCA-NP groups and the control. The iBCA-NPs caused no alteration in stress, oxidative stress (superoxide dismutase, SOD), plasma complement titer, or lysozyme activity. This study presents the first report of antibacterial activity of iBCA-NPs against Gram-negative bacteria. The results of this study suggest that D60 and NP30 may contribute to reducing the amounts of antibiotics and chemotherapeutic agents used in aquaculture.
Collapse
Affiliation(s)
- Mawuko G. Ahiable
- Laboratory of Cell Structure and Function, Division of Marine Bioresource Science, Graduate School of Kuroshio Science, Kochi University, Nankoku Kochi 783-8502, Japan; (M.G.A.); (K.M.); (M.H.)
| | - Kouki Matsunaga
- Laboratory of Cell Structure and Function, Division of Marine Bioresource Science, Graduate School of Kuroshio Science, Kochi University, Nankoku Kochi 783-8502, Japan; (M.G.A.); (K.M.); (M.H.)
| | - Mao Hokin
- Laboratory of Cell Structure and Function, Division of Marine Bioresource Science, Graduate School of Kuroshio Science, Kochi University, Nankoku Kochi 783-8502, Japan; (M.G.A.); (K.M.); (M.H.)
| | - Kazuhiro Iida
- Chikami Miltec Inc., 1-6-3 Ohtesuji, Kochi City 780-0842, Japan; (K.I.); (F.B.)
| | - Fumiaki Befu
- Chikami Miltec Inc., 1-6-3 Ohtesuji, Kochi City 780-0842, Japan; (K.I.); (F.B.)
| | - Syun-Ichirou Oshima
- Laboratory of Cell Structure and Function, Division of Marine Bioresource Science, Graduate School of Kuroshio Science, Kochi University, Nankoku Kochi 783-8502, Japan; (M.G.A.); (K.M.); (M.H.)
| |
Collapse
|
7
|
Sun B, Paraskevopoulos G, Min J, Rossdeutcher R, Ghosh S, Quinn B, Lin M, Sarkar D, Sukumaran D, Wang Y, Vávrová K, Lovell JF, Zhang Y. Topical Drug Delivery of Concentrated Cabazitaxel in an α-Tocopherol and DMSO Solution. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302658. [PMID: 37555802 PMCID: PMC10582425 DOI: 10.1002/advs.202302658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/03/2023] [Indexed: 08/10/2023]
Abstract
Topical chemotherapy approaches are relevant for certain skin cancer treatments. This study observes that cabazitaxel (CTX), a broad-spectrum second-generation taxane cytotoxic agent, can be dissolved in α-tocopherol at high concentrations exceeding 100 mg mL-1 . 2D nuclear magnetic resonance (NMR) analysis and molecular dynamics (MD) are used to study this phenomenon. The addition of 30% dimethyl sulfoxide (DMSO) to the α-tocopherol/CTX solution improves its working viscosity and enhances CTX permeation through human skin in vitro (over 5 µg cm-2 within 24 h), while no detectable drug permeates when CTX is dissolved in α-tocopherol alone. In a transepidermal water loss assay, the barrier impairment induced by CTX in 30% DMSO in α-tocopherol, but not in pure DMSO, is reversible 8 h after the formulation removal from the skin surface. Antitumor efficacy of the topical CTX formulation is evaluated in nude mice bearing A431 human squamous carcinoma skin cancer xenografts. With topical application of concentrated CTX solutions (75 mg mL-1 ), tumor growth is significantly suppressed compared to lower concentration groups (0, 25, or 50 mg mL-1 CTX). Taken together, these findings show that topical delivery of CTX using a DMSO and α-tocopherol solvent warrants further study as a treatment for skin malignancies.
Collapse
Affiliation(s)
- Boyang Sun
- School of Chemical Engineering and TechnologyKey Laboratory of Systems Bioengineering (Ministry of Education)Frontiers Science Center for Synthetic Biology (Ministry of Education)State Key Laboratory of Chemical EngineeringTianjin UniversityTianjin300350P. R. China
| | - Georgios Paraskevopoulos
- Skin Barrier Research GroupFaculty of PharmacyCharles UniversityAkademika Heyrovského 1203Hradec Králové50005Czech Republic
| | - Jiwei Min
- School of Chemical Engineering and TechnologyKey Laboratory of Systems Bioengineering (Ministry of Education)Frontiers Science Center for Synthetic Biology (Ministry of Education)State Key Laboratory of Chemical EngineeringTianjin UniversityTianjin300350P. R. China
| | - Robert Rossdeutcher
- Department of ChemistryState University of New York at BuffaloBuffaloNY14260USA
| | - Sanjana Ghosh
- Department of Biomedical EngineeringState University of New York at BuffaloBuffaloNY14260USA
| | - Breandan Quinn
- Department of Biomedical EngineeringState University of New York at BuffaloBuffaloNY14260USA
| | - Meng‐Hsuan Lin
- Department of Biomedical EngineeringState University of New York at BuffaloBuffaloNY14260USA
| | - Debanjan Sarkar
- Department of Biomedical EngineeringState University of New York at BuffaloBuffaloNY14260USA
| | - Dinesh Sukumaran
- Department of ChemistryState University of New York at BuffaloBuffaloNY14260USA
| | - Yuefei Wang
- School of Chemical Engineering and TechnologyKey Laboratory of Systems Bioengineering (Ministry of Education)Frontiers Science Center for Synthetic Biology (Ministry of Education)State Key Laboratory of Chemical EngineeringTianjin UniversityTianjin300350P. R. China
| | - Kateřina Vávrová
- Skin Barrier Research GroupFaculty of PharmacyCharles UniversityAkademika Heyrovského 1203Hradec Králové50005Czech Republic
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringState University of New York at BuffaloBuffaloNY14260USA
| | - Yumiao Zhang
- School of Chemical Engineering and TechnologyKey Laboratory of Systems Bioengineering (Ministry of Education)Frontiers Science Center for Synthetic Biology (Ministry of Education)State Key Laboratory of Chemical EngineeringTianjin UniversityTianjin300350P. R. China
| |
Collapse
|
8
|
Wang J, Xu P, Zhang Y, Han S, Wang G, Wang H, Song H, Li S. Dynamic nanoassemblies derived from small-molecule homodimeric prodrugs for in situ drug activation and safe osteosarcoma treatment. iScience 2023; 26:107409. [PMID: 37554455 PMCID: PMC10404730 DOI: 10.1016/j.isci.2023.107409] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/30/2023] [Accepted: 07/13/2023] [Indexed: 08/10/2023] Open
Abstract
Supramolecular prodrug self-assembly is a cost-effective and powerful approach for creating injectable anticancer nanoassemblies. Herein, we describe the self-assembly of small-molecule prodrug nanotherapeutics for tumor-restricted pharmacology that can be self-activated and independent of the exogenous stimuli. Covalent dimerization of the anticancer agent cabazitaxel via reactive oxygen species (ROS)- and esterase-activatable linkages produced the homodimeric prodrug diCTX, which was further coassembled with an ROS generator, dimeric dihydroartemisinin (diDHA). The coassembled nanoparticles were further refined in an amphiphilic matrix, making them suitable for in vivo administration. The ROS obtained from the coassembled diDHA synergized with intracellular esterase to activate the neighboring diCTX, which in turn induced potent cytotoxicity. In a preclinical orthotopic model of human osteosarcomas, nanoparticle administration exhibited durable antitumor efficacy. Furthermore, this smart, dual-responsive nanotherapeutic exhibited lower toxicity in animals than those of free drug combinations. We predict that this platform has great potential for further clinical translation.
Collapse
Affiliation(s)
- Jian Wang
- Department of Orthopedics, Shanghai Pudong New Area People’s Hospital, Shanghai, P.R. China
| | - Peirong Xu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province 250117, P.R. China
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, P.R. China
- Department of Chemical Engineering, Zhejiang University, Hangzhou, Zhejiang Province 310027, P.R. China
| | - Yeyong Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Shuai Han
- Department of Orthopedics, Shanghai Pudong New Area People’s Hospital, Shanghai, P.R. China
| | - Gongteng Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Hangxiang Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province 250117, P.R. China
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, P.R. China
| | - Haihan Song
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People’s Hospital, Shanghai, P.R. China
| | - Shufeng Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| |
Collapse
|
9
|
Pourmadadi M, Ghaemi A, Shaghaghi M, Rahdar A, Pandey S. Cabazitaxel-nano delivery systems as a cutting-edge for cancer therapy. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
10
|
Sun B, Lovell JF, Zhang Y. Current development of cabazitaxel drug delivery systems. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1854. [PMID: 36161272 DOI: 10.1002/wnan.1854] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/23/2022] [Accepted: 08/30/2022] [Indexed: 11/08/2022]
Abstract
The second-generation taxane cabazitaxel has been clinically approved for the treatment of metastatic castration-resistant prostate cancer after docetaxel failure. Compared with the first-generation taxanes paclitaxel and docetaxel, cabazitaxel has potent anticancer activity and is less prone to drug resistance due to its lower affinity for the P-gp efflux pump. The relatively high hydrophobicity of cabazitaxel and the poor aqueous colloidal stability of the commercial formulation, following its preparation for injection, presents opportunities for new cabazitaxel formulations with improved features. This review provides an overview of cabazitaxel drug formulations and hydrophobic taxane drug delivery systems in general, and particularly focuses on emerging cabazitaxel delivery systems discovered in the past 5 years. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Boyang Sun
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, China
| |
Collapse
|
11
|
Hyldbakk A, Fleten KG, Snipstad S, Åslund AKO, Davies CDL, Flatmark K, Mørch Y. Intraperitoneal administration of cabazitaxel-loaded nanoparticles in peritoneal metastasis models. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102656. [PMID: 36646195 DOI: 10.1016/j.nano.2023.102656] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/20/2022] [Accepted: 01/01/2023] [Indexed: 01/15/2023]
Abstract
Colorectal and ovarian cancers frequently develop peritoneal metastases with few treatment options. Intraperitoneal chemotherapy has shown promising therapeutic effects, but is limited by rapid drug clearance and systemic toxicity. We therefore encapsulated the cabazitaxel taxane in poly(alkyl cyanoacrylate) (PACA) nanoparticles (NPs), designed to improve intraperitoneal delivery. Toxicity of free and encapsulated cabazitaxel was investigated in rats by monitoring clinical signs, organ weight and blood hematological and biochemical parameters. Pharmacokinetics, biodistribution and treatment response were evaluated in mice. Biodistribution was investigated by measuring both cabazitaxel and the 2-ethylbutanol NP degradation product. Drug encapsulation was shown to increase intraperitoneal drug retention, leading to prolonged intraperitoneal drug residence time and higher drug concentrations in peritoneal tumors. As a result, encapsulation of cabazitaxel improved the treatment response in two in vivo models bearing intraperitoneal tumors. Together, these observations indicate a strong therapeutic potential of NP-based cabazitaxel encapsulation as a novel treatment for peritoneal metastases.
Collapse
Affiliation(s)
- Astrid Hyldbakk
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Karianne Giller Fleten
- Department of Tumor Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Sofie Snipstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway.
| | - Andreas K O Åslund
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway.
| | | | - Kjersti Flatmark
- Department of Tumor Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Gastroenterological Surgery, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway.
| |
Collapse
|
12
|
Hyldbakk A, Mørch Y, Snipstad S, Åslund AKO, Klinkenberg G, Nakstad VT, Wågbø AM, Schmid R, Molesworth PP. Identification of novel cyanoacrylate monomers for use in nanoparticle drug delivery systems prepared by miniemulsion polymerisation - A multistep screening approach. Int J Pharm X 2022; 4:100124. [PMID: 35898812 PMCID: PMC9310130 DOI: 10.1016/j.ijpx.2022.100124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/24/2022] Open
Abstract
Poly (alkyl cyanoacrylate) (PACA) polymeric nanoparticles (NPs) are promising drug carriers in drug delivery. However, the selection of commercially available alkyl cyanoacrylate (ACA) monomers is limited, because most monomers were designed for use in medical and industrial glues and later repurposed for drug encapsulation. This study therefore aimed to seek out novel ACA materials for use in NP systems using a toxicity led screening approach. A multistep strategy, including cytotoxicity screening of alcohols as degradation products of PACA (44 alcohols), NPs (14 polymers), and a final in vivo study (2 polymers) gave poly (2-ethylhexyl cyanoacrylate) PEHCA as a promising novel PACA candidate. For the first time, this work presents cytotoxicity data on several novel ACAs, PEHCA in vivo toxicity data, and miniemulsion polymerisation-based encapsulation of the cabazitaxel and NR688 in novel PACA candidates. Furthermore, several of the ACA candidates were compatible with a wider selection of lipophilic active pharmaceutical ingredients (APIs) versus commercially available controls. Combined, this work demonstrates the potential benefits of expanding the array of available ACA materials in drug delivery. Novel ACAs have the potential to encapsulate a wider range of APIs in miniemulsion polymerisation processes and may also broaden PACA applicability in other fields. Screening of novel poly(alkylcyanoacrylate) (PACA) materials to broaden PACA nanomedicine potential. A comprehensive screening process evaluated the toxicity of novel poly(alkylcyanoacrylate) (PACA) materials. Novel poly(2-ethylhexyl cyanoacrylate) nanoparticles has a promising safety profile. Novel ACA materials show potential to enable encapsulation of a wider range of APIs.
Collapse
Affiliation(s)
- Astrid Hyldbakk
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway.,Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Sofie Snipstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway.,Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway.,Cancer Clinic, St. Olavs Hospital, Trondheim, Norway
| | - Andreas K O Åslund
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Geir Klinkenberg
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Vu To Nakstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Ane-Marit Wågbø
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Ruth Schmid
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Peter P Molesworth
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| |
Collapse
|
13
|
Rodrigues Arruda B, Mendes MGA, Freitas PGCD, Reis AVF, Lima T, Crisóstomo LCCF, Nogueira KAB, Pessoa C, Petrilli R, Eloy JO. Nanocarriers for delivery of taxanes: A review on physicochemical and biological aspects. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14
|
Fleten KG, Hyldbakk A, Einen C, Benjakul S, Strand BL, Davies CDL, Mørch Ý, Flatmark K. Alginate Microsphere Encapsulation of Drug-Loaded Nanoparticles: A Novel Strategy for Intraperitoneal Drug Delivery. Mar Drugs 2022; 20:744. [PMID: 36547891 PMCID: PMC9782800 DOI: 10.3390/md20120744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Alginate hydrogels have been broadly investigated for use in medical applications due to their biocompatibility and the possibility to encapsulate cells, proteins, and drugs. In the treatment of peritoneal metastasis, rapid drug clearance from the peritoneal cavity is a major challenge. Aiming to delay drug absorption and reduce toxic side effects, cabazitaxel (CAB)-loaded poly(alkyl cyanoacrylate) (PACA) nanoparticles were encapsulated in alginate microspheres. The PACAlg alginate microspheres were synthesized by electrostatic droplet generation and the physicochemical properties, stability, drug release kinetics, and mesothelial cytotoxicity were analyzed before biodistribution and therapeutic efficacy were studied in mice. The 450 µm microspheres were stable at in vivo conditions for at least 21 days after intraperitoneal implantation in mice, and distributed evenly throughout the peritoneal cavity without aggregation or adhesion. The nanoparticles were stably retained in the alginate microspheres, and nanoparticle toxicity to mesothelial cells was reduced, while the therapeutic efficacy of free CAB was maintained or improved in vivo. Altogether, this work presents the alginate encapsulation of drug-loaded nanoparticles as a promising novel strategy for the treatment of peritoneal metastasis that can improve the therapeutic ratio between toxicity and therapeutic efficacy.
Collapse
Affiliation(s)
- Karianne Giller Fleten
- Department of Tumor Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Astrid Hyldbakk
- Department of Biotechnology and Nanomedicine, SINTEF Industry, 7034 Trondheim, Norway
- Department of Physics, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Caroline Einen
- Department of Physics, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Sopisa Benjakul
- Department of Physics, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Berit Løkensgard Strand
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Catharina de Lange Davies
- Department of Physics, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, 7034 Trondheim, Norway
| | - Kjersti Flatmark
- Department of Tumor Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Department of Gastroenterological Surgery, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| |
Collapse
|
15
|
Pourmadadi M, Rahmani E, Shamsabadipour A, Mahtabian S, Ahmadi M, Rahdar A, Díez-Pascual AM. Role of Iron Oxide (Fe 2O 3) Nanocomposites in Advanced Biomedical Applications: A State-of-the-Art Review. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3873. [PMID: 36364649 PMCID: PMC9653814 DOI: 10.3390/nano12213873] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 06/16/2023]
Abstract
Nanomaterials have demonstrated a wide range of applications and recently, novel biomedical studies are devoted to improving the functionality and effectivity of traditional and unmodified systems, either drug carriers and common scaffolds for tissue engineering or advanced hydrogels for wound healing purposes. In this regard, metal oxide nanoparticles show great potential as versatile tools in biomedical science. In particular, iron oxide nanoparticles with different shape and sizes hold outstanding physiochemical characteristics, such as high specific area and porous structure that make them idoneous nanomaterials to be used in diverse aspects of medicine and biological systems. Moreover, due to the high thermal stability and mechanical strength of Fe2O3, they have been combined with several polymers and employed for various nano-treatments for specific human diseases. This review is focused on summarizing the applications of Fe2O3-based nanocomposites in the biomedical field, including nanocarriers for drug delivery, tissue engineering, and wound healing. Additionally, their structure, magnetic properties, biocompatibility, and toxicity will be discussed.
Collapse
Affiliation(s)
- Mehrab Pourmadadi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 14174, Iran
| | - Erfan Rahmani
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 14174, Iran
| | - Amin Shamsabadipour
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 14174, Iran
| | - Shima Mahtabian
- Department of Materials Engineering, Shahreza Bramch, Islamic Azad University, Shahreza, Isfahan 61349-37333, Iran
| | - Mohammadjavad Ahmadi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 14174, Iran
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol 538-98615, Iran
| | - Ana M. Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain
| |
Collapse
|
16
|
Åslund AKO, Vandebriel RJ, Caputo F, de Jong WH, Delmaar C, Hyldbakk A, Rustique E, Schmid R, Snipstad S, Texier I, Vernstad K, Borgos SEF. A comparative biodistribution study of polymeric and lipid-based nanoparticles. Drug Deliv Transl Res 2022; 12:2114-2131. [PMID: 35426570 PMCID: PMC9012159 DOI: 10.1007/s13346-022-01157-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 12/24/2022]
Abstract
Biodistribution of nanoencapsulated bioactive compounds is primarily determined by the size, shape, chemical composition and surface properties of the encapsulating nanoparticle, and, thus, less dependent on the physicochemical properties of the active pharmaceutical ingredient encapsulated. In the current work, we aimed to investigate the impact of formulation type on biodistribution profile for two clinically relevant nanoformulations. We performed a comparative study of biodistribution in healthy rats at several dose levels and durations up to 14-day post-injection. The studied nanoformulations were nanostructured lipid carriers incorporating the fluorescent dye IR780-oleyl, and polymeric nanoparticles containing the anticancer agent cabazitaxel. The biodistribution was approximated by quantification of the cargo in blood and relevant organs. Several clear and systematic differences in biodistribution were observed, with the most pronounced being a much higher (more than 50-fold) measured concentration ratio between cabazitaxel in all organs vs. blood, as compared to IR780-oleyl. Normalized dose linearity largely showed opposite trends between the two compounds after injection. Cabazitaxel showed a higher brain accumulation than IR780-oleyl with increasing dose injected. Interestingly, cabazitaxel showed a notable and prolonged accumulation in lung tissue compared to other organs. The latter observations could warrant further studies towards a possible therapeutic indication within lung and conceivably brain cancer for nanoformulations of this highly antineoplastic compound, for which off-target toxicity is currently dose-limiting in the clinic.
Collapse
|
17
|
Sønstevold T, Engedal N, Torgersen ML. Perturbation of Cellular Redox Homeostasis Dictates Divergent Effects of Polybutyl Cyanoacrylate (PBCA) Nanoparticles on Autophagy. Cells 2021; 10:3432. [PMID: 34943939 PMCID: PMC8699722 DOI: 10.3390/cells10123432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/20/2021] [Accepted: 12/03/2021] [Indexed: 01/18/2023] Open
Abstract
Nanoparticles (NPs) are used in our everyday life, including as drug delivery vehicles. However, the effects of NPs at the cellular level and their impacts on autophagy are poorly understood. Here, we demonstrate that the NP drug delivery vehicle poly(butyl cyanoacrylate) (PBCA) perturbs redox homeostasis in human epithelial cells, and that the degree of redox perturbation dictates divergent effects of PBCA on autophagy. Specifically, PBCA promoted functional autophagy at low concentrations, whereas it inhibited autophagy at high concentrations. Both effects were completely abolished by the antioxidant N-acetyl cysteine (NAC). High concentrations of PBCA inhibited MAP1LC3B/GABARAP lipidation and LC3 flux, and blocked bulk autophagic cargo flux induced by mTOR inhibition. These effects were mimicked by the redox regulator H2O2. In contrast, low concentrations of PBCA enhanced bulk autophagic cargo flux in a Vps34-, ULK1/2- and ATG13-dependent manner, yet interestingly, without an accompanying increase in LC3 lipidation or flux. PBCA activated MAP kinase signaling cascades in a redox-dependent manner, and interference with individual signaling components revealed that the autophagy-stimulating effect of PBCA required the action of the JNK and p38-MK2 pathways, whose activities converged on the pro-autophagic protein Beclin-1. Collectively, our results reveal that PBCA exerts a dual effect on autophagy depending on the severity of the NP insult and the resulting perturbation of redox homeostasis. Such a dual autophagy-modifying effect may be of general relevance for redox-perturbing NPs and have important implications in nanomedicine.
Collapse
Affiliation(s)
- Tonje Sønstevold
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway;
| | - Nikolai Engedal
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway;
| | - Maria Lyngaas Torgersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway;
| |
Collapse
|
18
|
Ahmadi M, Pourmadadi M, Ghorbanian SA, Yazdian F, Rashedi H. Ultra pH-sensitive nanocarrier based on Fe 2O 3/chitosan/montmorillonite for quercetin delivery. Int J Biol Macromol 2021; 191:738-745. [PMID: 34517028 DOI: 10.1016/j.ijbiomac.2021.09.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/20/2021] [Accepted: 09/04/2021] [Indexed: 02/06/2023]
Abstract
Harmful side effects of the chemotherapeutic agent have been investigated in many recent studies. Since Fe2O3 nanoparticles have proper porosity, they are capable for loading noticeable amount of drugs and controlled release. We developed Fe2O3/chitosan/montmorillonite nanocomposite. Quercetin (QC) nanoparticles, which have fewer side effects than chemical anti-tumor drugs, were encapsulated in the synthesized nanocarrier and were characterized by X-ray diffraction (XRD), Fourier transforms infrared spectroscopy (FTIR), field emission scanning electron microscopy (FE-SEM), vibrating sample magnetometer (VSM), dynamic light scattering (DLS), and zeta potential. For quercetin, the encapsulation efficiency and the loading efficiency of the drug in Fe2O3-CS-MMT@QC were found to be about 94% and 57%, respectively. The release profile of QC in different mediums indicated pH-dependency and controlled release of the nanocomposite, adhering to The Weibull kinetic model. Biocompatibility of the Fe2O3/CS/MMT nanoparticles against the MCF-7 cells was shown by MTT assay and confirmed by flow cytometry. These data demonstrate that the designed Fe2O3-CS-MMT@QC would have potential drug delivery to treat cancer cells.
Collapse
Affiliation(s)
- Mohammadjavad Ahmadi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mehrab Pourmadadi
- Department of Life Science Engineering, Faculty of New Science and Technology, University of Tehran, Iran
| | - Sohrab Ali Ghorbanian
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| | - Fatemeh Yazdian
- Department of Life Science Engineering, Faculty of New Science and Technology, University of Tehran, Iran.
| | - Hamid Rashedi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| |
Collapse
|
19
|
Qu Z, Ren Y, Shen H, Wang H, Shi L, Tong D. Combination Therapy of Metastatic Castration-Recurrent Prostate Cancer: Hyaluronic Acid Decorated, Cabazitaxel-Prodrug and Orlistat Co-Loaded Nano-System. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3605-3616. [PMID: 34447241 PMCID: PMC8384126 DOI: 10.2147/dddt.s306684] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/03/2021] [Indexed: 12/29/2022]
Abstract
Purpose Prostate cancer (PCa) is the second leading cause of cancer-related death among men in developed countries. Cabazitaxel (CBZ) is recommended as one of the most active chemotherapy agents for PCa. This study aimed to develop a hyaluronic acid (HA) decorated, cabazitaxel-prodrug (HA-CBZ) and orlistat (ORL) co-loaded nano-system against the prostate cancer in vitro and in vivo. Methods Cabazitaxel-prodrug was firstly synthesized by conjugating HA with CBZ through the formation of ester bonds. HA contained ORL and CBZ prodrug co-loaded lipid-polymer hybrid nanoparticles (ORL/HA-CBZ/LPNs) were constructed and characterized in terms of particle size, zeta potential, drug loading capacity and stability. The antitumor efficiency and systemic toxicity of LPNs were evaluated in vitro and in vivo. Results The resulting ORL/HA-CBZ/LPNs were 150.9 nm in particle size with narrow distribution and high entrapment efficiency. The minimum combination index of 0.57 was found at a drug ratio of 1:2 (ORL:HA-CBZ, w/w) in the drug co-loaded formulations, indicating the strongest synergism effect. ORL/HA-CBZ/LPNs demonstrated an enhanced in vitro and in vivo antitumor effect compared with single drug loaded LPNs and free drug formulations. Conclusion ORL/HA-CBZ/LPNs showed remarkable synergism cytotoxicity and the best tumor inhibition efficiency in mice with negligible systemic toxicity. ORL/HA-CBZ/LPNs can be highly useful for targeted prostate cancer therapy.
Collapse
Affiliation(s)
- Zhen Qu
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| | - Yuning Ren
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| | - Hongyu Shen
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| | - Huihui Wang
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| | - Lijie Shi
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| | - Deyong Tong
- Department of Oncology, 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264001, People's Republic of China
| |
Collapse
|
20
|
de Souza ML, de Albuquerque Wanderley Sales V, Alves L, Santos WM, Ferraz LR, Lima G, Mendes L, Rolim LA, Neto PJR. A systematic review of functionalized polymeric nanoparticles to improve intestinal permeability of drugs and biological products. Curr Pharm Des 2021; 28:410-426. [PMID: 34348618 DOI: 10.2174/1381612827666210804104205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/19/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The oral route is the most frequently used and the most convenient route of drug administration, since it has several advantages, such as ease of use, patient compliance and better cost-effectiveness. However, physicochemical and biopharmaceutical limitations of various active pharmaceutical ingredients (API) hinder suitability for this route, including degradation in the gastrointestinal tract, low intestinal permeability and low bioavailability. To overcome these problems, while maintaining therapeutic efficacy, polymeric nanoparticles have attracted considerable attention for their ability to increase drug solubility, promote controlled release, and improve stability. In addition, the functionalization of nanocarriers can increase uptake and accumulation at the target site of action, and intestinal absorption, making it possible to obtain more viable, safe and efficient treatments for oral administration. <P> Objective: This systematic review aimed to seek recent advances in the literature on the use of polymeric nanoparticles functionalization to increase intestinal permeability of APIs that are intended for oral administration. <P> Method: Two bibliographic databases were consulted (PubMed and ScienceDirect). The selected publications and the writing of this systematic review were based on the guidelines mentioned in the PRISMA statement. <P> Results: Out of a total of 3036 studies, 22 studies were included in this article based on our eligibility criteria. The results were consistent for the application of nanoparticle functionalization to increase intestinal permeability. <P> Conclusion: The functionalized polymeric nanoparticles can be considered as carrier systems that improve the intestinal permeability and bioavailability of APIs, with the potential to result, in the future, in the development of oral medicines.
Collapse
Affiliation(s)
- Myla Lôbo de Souza
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | | | - Larissa Alves
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | - Widson Michael Santos
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | - Leslie Raphael Ferraz
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | - Gustavo Lima
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | - Larissa Mendes
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | - Larissa Araújo Rolim
- Central de Análise de Fármacos, Medicamentos e Alimentos. Federal University of Vale do São Francisco (UNIVASF), Petrolina-PE. Brazil
| | - Pedro José Rolim Neto
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| |
Collapse
|
21
|
Sulheim E, Hanson I, Snipstad S, Vikedal K, Mørch Y, Boucher Y, Davies CDL. Sonopermeation with Nanoparticle‐Stabilized Microbubbles Reduces Solid Stress and Improves Nanomedicine Delivery to Tumors. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Einar Sulheim
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
- Cancer Clinic St.Olavs Hospital Trondheim 7030 Norway
| | - Ingunn Hanson
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| | - Sofie Snipstad
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
- Cancer Clinic St.Olavs Hospital Trondheim 7030 Norway
| | - Krister Vikedal
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
| | - Yves Boucher
- Edwin L. Steele Laboratory for Tumor Biology Massachusetts General Hospital Boston MA 02114 USA
| | - Catharina de Lange Davies
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| |
Collapse
|
22
|
Niza E, Ocaña A, Castro-Osma JA, Bravo I, Alonso-Moreno C. Polyester Polymeric Nanoparticles as Platforms in the Development of Novel Nanomedicines for Cancer Treatment. Cancers (Basel) 2021; 13:3387. [PMID: 34298604 PMCID: PMC8304499 DOI: 10.3390/cancers13143387] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/24/2021] [Accepted: 06/29/2021] [Indexed: 12/16/2022] Open
Abstract
Many therapeutic agents have failed in their clinical development, due to the toxic effects associated with non-transformed tissues. In this context, nanotechnology has been exploited to overcome such limitations, and also improve navigation across biological barriers. Amongst the many materials used in nanomedicine, with promising properties as therapeutic carriers, the following one stands out: biodegradable and biocompatible polymers. Polymeric nanoparticles are ideal candidates for drug delivery, given the versatility of raw materials and their feasibility in large-scale production. Furthermore, polymeric nanoparticles show great potential for easy surface modifications to optimize pharmacokinetics, including the half-life in circulation and targeted tissue delivery. Herein, we provide an overview of the current applications of polymeric nanoparticles as platforms in the development of novel nanomedicines for cancer treatment. In particular, we will focus on the raw materials that are widely used for polymeric nanoparticle generation, current methods for formulation, mechanism of action, and clinical investigations.
Collapse
Affiliation(s)
- Enrique Niza
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain; (E.N.); (J.A.C.-O.)
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| | - Alberto Ocaña
- Experimental Therapeutics Unit, Hospital Clínico San Carlos, IdISSC and CIBERONC, 28040 Madrid, Spain;
| | - José Antonio Castro-Osma
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain; (E.N.); (J.A.C.-O.)
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| | - Iván Bravo
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain; (E.N.); (J.A.C.-O.)
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| | - Carlos Alonso-Moreno
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain; (E.N.); (J.A.C.-O.)
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| |
Collapse
|
23
|
Øverbye A, Torgersen ML, Sønstevold T, Iversen TG, Mørch Ý, Skotland T, Sandvig K. Cabazitaxel-loaded poly(alkyl cyanoacrylate) nanoparticles: toxicity and changes in the proteome of breast, colon and prostate cancer cells. Nanotoxicology 2021; 15:865-884. [PMID: 34047629 DOI: 10.1080/17435390.2021.1924888] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Nanoparticles composed of poly(alkyl cyanoacrylate) (PACA) have shown great promise due to their biodegradability and high drug loading capacity. Development of optimal PACA nanocarriers requires detailed analysis of the overall cellular impact exerted by PACA variants. We here perform a comprehensive comparison of cabazitaxel (CBZ)-loaded nanocarriers composed of three different PACA monomers, i.e. poly(n-butyl cyanoacrylate) (PBCA), poly(2-ethylbutyl cyanoacrylate) (PEBCA) and poly(octyl cyanoacrylate) (POCA). The cytotoxicity of drug-loaded and empty PACA nanoparticles were compared to that of free CBZ across a panel of nine cancer cell lines by assessing cellular metabolism, proliferation and protein synthesis. The analyses revealed that the cytotoxicity of all CBZ-loaded PACAs was similar to that of free CBZ for all cell lines tested, whereas the empty PACAs exerted much lower toxicity. To increase our understanding of the toxic effects of these treatments comprehensive MS-based proteomics were performed with HCT116, MDA-MB-231 and PC3 cells incubated with PACA-CBZ variants or free CBZ. Interestingly, PACA-CBZ specifically led to decreased levels of proteins involved in focal adhesion and stress fibers in all cell lines. Since we recently demonstrated that encapsulation of CBZ within PEBCA nanoparticles significantly improved the therapeutic effect of CBZ on a patient derived xenograft model in mice, we investigated the effects of this PACA variant more closely by immunoblotting. Interestingly, we detected several changes in the protein expression and degree of phosphorylation of SRC-pathway proteins that can be relevant for the therapeutic effects of these substances.
Collapse
Affiliation(s)
- Anders Øverbye
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Maria Lyngaas Torgersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Tonje Sønstevold
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Tore Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
24
|
Snipstad S, Mørch Ý, Sulheim E, Åslund A, Pedersen A, Davies CDL, Hansen R, Berg S. Sonopermeation Enhances Uptake and Therapeutic Effect of Free and Encapsulated Cabazitaxel. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1319-1333. [PMID: 33549379 DOI: 10.1016/j.ultrasmedbio.2020.12.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/18/2020] [Accepted: 12/27/2020] [Indexed: 06/12/2023]
Abstract
Delivery of drugs and nanomedicines to tumors is often heterogeneous and insufficient and, thus, of limited efficacy. Microbubbles in combination with ultrasound have been found to improve delivery to tumors, enhancing accumulation and penetration. We used a subcutaneous prostate cancer xenograft model in mice to investigate the effect of free and nanoparticle-encapsulated cabazitaxel in combination with ultrasound and microbubbles with a lipid shell or a shell of nanoparticles. Sonopermeation reduced tumor growth and prolonged survival (26%-100%), whether the free drug was co-injected with lipid-shelled microbubbles or the nanoformulation was co-injected with lipid-shelled or nanoparticle-shelled microbubbles. Coherently with the improved therapeutic response, we found enhanced uptake of nanoparticles directly after ultrasound treatment that lasted several weeks (2.3 × -15.8 × increase). Neither cavitation dose nor total accumulation of nanoparticles could explain the variation within treatment groups, emphasizing the need for a better understanding of the tumor biology and mechanisms involved in ultrasound-mediated treatment.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway.
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Einar Sulheim
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Andreas Åslund
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - André Pedersen
- Department of Health Research, SINTEF Digital, Trondheim, Norway
| | | | - Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sigrid Berg
- Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical imaging, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
25
|
Pandya AD, Iversen TG, Moestue S, Grinde MT, Mørch Ý, Snipstad S, Åslund AKO, Øy GF, Kildal W, Engebråten O, Sandvig K, Skotland T, Mælandsmo GM. Biodistribution of Poly(alkyl cyanoacrylate) Nanoparticles in Mice and Effect on Tumor Infiltration of Macrophages into a Patient-Derived Breast Cancer Xenograft. NANOMATERIALS 2021; 11:nano11051140. [PMID: 33924869 PMCID: PMC8145722 DOI: 10.3390/nano11051140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 11/18/2022]
Abstract
We have investigated the biodistribution and tumor macrophage infiltration after intravenous injection of the poly(alkyl cyanoacrylate) nanoparticles (NPs): PEBCA (poly(2-ethyl-butyl cyanoacrylate), PBCA (poly(n-butyl cyanoacrylate), and POCA (poly(octyl cyanoacrylate), in mice. These NPs are structurally similar, have similar PEGylation, and have previously been shown to give large variations in cellular responses in vitro. The PEBCA NPs had the highest uptake both in the patient-derived breast cancer xenograft MAS98.12 and in lymph nodes, and therefore, they are the most promising of these NPs for delivery of cancer drugs. High-resolution magic angle spinning magnetic resonance (HR MAS MR) spectroscopy did not reveal any differences in the metabolic profiles of tumors following injection of the NPs, but the PEBCA NPs resulted in higher tumor infiltration of the anti-tumorigenic M1 macrophages than obtained with the two other NPs. The PEBCA NPs also increased the ratio of M1/M2 (anti-tumorigenic/pro-tumorigenic) macrophages in the tumors, suggesting that these NPs might be used both as a vehicle for drug delivery and to modulate the immune response in favor of enhanced therapeutic effects.
Collapse
Affiliation(s)
- Abhilash D. Pandya
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (A.D.P.); (G.F.Ø.); (O.E.)
| | - Tore-Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (T.-G.I.); (K.S.)
| | - Siver Moestue
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway;
| | - Maria T. Grinde
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, 7491 Trondheim, Norway;
| | - Ýrr Mørch
- SINTEF AS, Department of Biotechnology and Nanomedicine, 7034 Trondheim, Norway; (Ý.M.); (S.S.); (A.K.O.Å.)
| | - Sofie Snipstad
- SINTEF AS, Department of Biotechnology and Nanomedicine, 7034 Trondheim, Norway; (Ý.M.); (S.S.); (A.K.O.Å.)
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Cancer Clinic, St. Olav’s Hospital, 7030 Trondheim, Norway
| | - Andreas K. O. Åslund
- SINTEF AS, Department of Biotechnology and Nanomedicine, 7034 Trondheim, Norway; (Ý.M.); (S.S.); (A.K.O.Å.)
| | - Geir F. Øy
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (A.D.P.); (G.F.Ø.); (O.E.)
| | - Wanja Kildal
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway;
| | - Olav Engebråten
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (A.D.P.); (G.F.Ø.); (O.E.)
- Department of Oncology, Oslo University Hospital, 0450 Oslo, Norway
- Institute of Clinical Medicine, Medical Faculty, University of Oslo, 0315 Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (T.-G.I.); (K.S.)
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (T.-G.I.); (K.S.)
- Correspondence: (T.S.); (G.M.M.)
| | - Gunhild M. Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; (A.D.P.); (G.F.Ø.); (O.E.)
- Department of Medical Biology, University of Tromsø, 9019 Tromsø, Norway
- Correspondence: (T.S.); (G.M.M.)
| |
Collapse
|
26
|
Song S, Xia H, Guo M, Wang S, Zhang S, Ma P, Jin Y. Role of macrophage in nanomedicine-based disease treatment. Drug Deliv 2021; 28:752-766. [PMID: 33860719 PMCID: PMC8079019 DOI: 10.1080/10717544.2021.1909175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Macrophages are a major component of the immunoresponse. Diversity and plasticity are two of the hallmarks of macrophages, which allow them to act as proinflammatory, anti-inflammatory, and homeostatic agents. Research has found that cancer and many inflammatory or autoimmune disorders are correlated with activation and tissue infiltration of macrophages. Recent developments in macrophage nanomedicine-based disease treatment are proving to be timely owing to the increasing inadequacy of traditional treatment. Here, we review the role of macrophages in nanomedicine-based disease treatment. First, we present a brief background on macrophages and nanomedicine. Then, we delve into applications of macrophages as a target for disease treatment and delivery systems and summarize the applications of macrophage-derived extracellular vesicles. Finally, we provide an outlook on the clinical utility of macrophages in nanomedicine-based disease treatment.
Collapse
Affiliation(s)
- Siwei Song
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Xia
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sufei Wang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujing Zhang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Hammond SM, Aartsma‐Rus A, Alves S, Borgos SE, Buijsen RAM, Collin RWJ, Covello G, Denti MA, Desviat LR, Echevarría L, Foged C, Gaina G, Garanto A, Goyenvalle AT, Guzowska M, Holodnuka I, Jones DR, Krause S, Lehto T, Montolio M, Van Roon‐Mom W, Arechavala‐Gomeza V. Delivery of oligonucleotide-based therapeutics: challenges and opportunities. EMBO Mol Med 2021; 13:e13243. [PMID: 33821570 PMCID: PMC8033518 DOI: 10.15252/emmm.202013243] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Nucleic acid-based therapeutics that regulate gene expression have been developed towards clinical use at a steady pace for several decades, but in recent years the field has been accelerating. To date, there are 11 marketed products based on antisense oligonucleotides, aptamers and small interfering RNAs, and many others are in the pipeline for both academia and industry. A major technology trigger for this development has been progress in oligonucleotide chemistry to improve the drug properties and reduce cost of goods, but the main hurdle for the application to a wider range of disorders is delivery to target tissues. The adoption of delivery technologies, such as conjugates or nanoparticles, has been a game changer for many therapeutic indications, but many others are still awaiting their eureka moment. Here, we cover the variety of methods developed to deliver nucleic acid-based therapeutics across biological barriers and the model systems used to test them. We discuss important safety considerations and regulatory requirements for synthetic oligonucleotide chemistries and the hurdles for translating laboratory breakthroughs to the clinic. Recent advances in the delivery of nucleic acid-based therapeutics and in the development of model systems, as well as safety considerations and regulatory requirements for synthetic oligonucleotide chemistries are discussed in this review on oligonucleotide-based therapeutics.
Collapse
Affiliation(s)
| | | | - Sandra Alves
- Department of Human Genetics, Research and Development UnitNational Health Institute Doutor Ricardo JorgePortoPortugal
| | - Sven E Borgos
- Department of Biotechnology and NanomedicineSINTEF ASTrondheimNorway
| | - Ronald A M Buijsen
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Rob W J Collin
- Department of Human Genetics and Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | - Giuseppina Covello
- Department of BiologyUniversity of PadovaPadovaItaly
- Department of Cellular, Computational and Integrative Biology ‐ CIBIOUniversity of TrentoTrentoItaly
| | - Michela A Denti
- Department of Cellular, Computational and Integrative Biology ‐ CIBIOUniversity of TrentoTrentoItaly
| | - Lourdes R Desviat
- Centro de Biología Molecular Severo Ochoa UAM‐CSICCIBERER, IdiPazUniversidad Autónoma de MadridMadridSpain
| | | | - Camilla Foged
- Department of PharmacyFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagen ØDenmark
| | - Gisela Gaina
- Victor Babes National Institute of PathologyBucharestRomania
- Department of Biochemistry and Molecular BiologyUniversity of BucharestBucharestRomania
| | - Alejandro Garanto
- Department of Human Genetics and Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
- Department of PediatricsRadboud University Medical CenterNijmegenThe Netherlands
| | | | - Magdalena Guzowska
- Department of Physiological SciencesFaculty of Veterinary MedicineWarsaw University of Life Sciences – SGGWWarsawPoland
| | - Irina Holodnuka
- Institute of Microbiology and VirologyRiga Stradins UniversityRigaLatvia
| | | | - Sabine Krause
- Department of NeurologyFriedrich‐Baur‐InstituteLudwig‐Maximilians‐University of MunichMunichGermany
| | - Taavi Lehto
- Institute of TechnologyUniversity of TartuTartuEstonia
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstitutetHuddingeSweden
| | - Marisol Montolio
- Duchenne Parent Project EspañaMadridSpain
- Department of Cell Biology, Fisiology and ImmunologyFaculty of BiologyUniversity of BarcelonaBarcelonaSpain
| | - Willeke Van Roon‐Mom
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Virginia Arechavala‐Gomeza
- Neuromuscular Disorders GroupBiocruces Bizkaia Health Research InstituteBarakaldoSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
| |
Collapse
|
28
|
Tieu T, Irani S, Bremert KL, Ryan NK, Wojnilowicz M, Helm M, Thissen H, Voelcker NH, Butler LM, Cifuentes‐Rius A. Patient-Derived Prostate Cancer Explants: A Clinically Relevant Model to Assess siRNA-Based Nanomedicines. Adv Healthc Mater 2021; 10:e2001594. [PMID: 33274851 DOI: 10.1002/adhm.202001594] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/05/2020] [Indexed: 01/27/2023]
Abstract
Over the last thirty years, research in nanomedicine has widely been focused on applications in cancer therapeutics. However, despite the plethora of reported nanoscale drug delivery systems that can successfully eradicate solid tumor xenografts in vivo, many of these formulations have not yet achieved clinical translation. This issue particularly pertains to the delivery of small interfering RNA (siRNA), a highly attractive tool for selective gene targeting. One of the likely reasons behind the lack of translation is that current in vivo models fail to recapitulate critical elements of clinical solid tumors that may influence drug response, such as cellular heterogeneity in the tumor microenvironment. This study incorporates a more clinically relevant model for assessing siRNA delivery systems; ex vivo culture of prostate cancer harvested from patients who have undergone radical prostatectomy, denoted patient-derived explants (PDE). The model retains native human tissue architecture, microenvironment, and cell signaling pathways. Porous silicon nanoparticles (pSiNPs) behavior in this model is investigated and compared with commonly used 3D cancer cell spheroids for their efficacy in the delivery of siRNA directed against the androgen receptor (AR), a key driver of prostate cancer.
Collapse
Affiliation(s)
- Terence Tieu
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Swati Irani
- South Australian Health & Medical Research Institute Adelaide SA 5001 Australia
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health University of Adelaide Adelaide SA 5005 Australia
| | - Kayla L. Bremert
- South Australian Health & Medical Research Institute Adelaide SA 5001 Australia
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health University of Adelaide Adelaide SA 5005 Australia
| | - Natalie K. Ryan
- South Australian Health & Medical Research Institute Adelaide SA 5001 Australia
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health University of Adelaide Adelaide SA 5005 Australia
| | | | - Madison Helm
- South Australian Health & Medical Research Institute Adelaide SA 5001 Australia
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health University of Adelaide Adelaide SA 5005 Australia
| | - Helmut Thissen
- CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton Victoria 3168 Australia
| | - Lisa M. Butler
- South Australian Health & Medical Research Institute Adelaide SA 5001 Australia
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health University of Adelaide Adelaide SA 5005 Australia
| | - Anna Cifuentes‐Rius
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
| |
Collapse
|
29
|
Fagerland SMT, Berg S, Hill DK, Snipstad S, Sulheim E, Hyldbakk A, Kim J, Davies CDL. Ultrasound-Mediated Delivery of Chemotherapy into the Transgenic Adenocarcinoma of the Mouse Prostate Model. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:3032-3045. [PMID: 32800470 DOI: 10.1016/j.ultrasmedbio.2020.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/19/2020] [Accepted: 07/07/2020] [Indexed: 06/11/2023]
Abstract
Ultrasound (US) in combination with microbubbles (MB) has had promising results in improving delivery of chemotherapeutic agents. However, most studies are done in immunodeficient mice with xenografted tumors. We used two phenotypes of the spontaneous transgenic adenocarcinoma of the mouse prostate (TRAMP) model to evaluate if US + MB could enhance the therapeutic efficacy of cabazitaxel (Cab). Cab was either injected intravenously as free drug or encapsulated into nanoparticles. In both cases, Cab transiently reduced tumor and prostate volume in the TRAMP model. No additional therapeutic efficacy was observed combining Cab with US + MB, except for one tumor. Additionally, histology grading and immunostaining of Ki67 did not reveal differences between treatment groups. Mass spectrometry revealed that nanoparticle encapsulation of Cab increased the circulation time and enhanced the accumulation in liver and spleen compared with free Cab. The therapeutic results in this spontaneous, clinically relevant tumor model differ from the improved therapeutic response observed in xenografts combining US + MB and chemotherapy.
Collapse
Affiliation(s)
- Stein-Martin T Fagerland
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sigrid Berg
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Deborah K Hill
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Astrid Hyldbakk
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Jana Kim
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | | |
Collapse
|
30
|
Juan A, Cimas FJ, Bravo I, Pandiella A, Ocaña A, Alonso-Moreno C. An Overview of Antibody Conjugated Polymeric Nanoparticles for Breast Cancer Therapy. Pharmaceutics 2020; 12:pharmaceutics12090802. [PMID: 32854255 PMCID: PMC7558516 DOI: 10.3390/pharmaceutics12090802] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 01/09/2023] Open
Abstract
Nanoparticles (NPs) are promising drug delivery systems (DDS) for identifying and treating cancer. Active targeting NPs can be generated by conjugation with ligands that bind overexpressed or mutant cell surface receptors on target cells that are poorly or not even expressed on normal cells. Receptor-mediated endocytosis of the NPs occurs and the drug is released inside the cell or in the surrounding tissue due to the bystander effect. Antibodies are the most frequently used ligands to actively target tumor cells. In this context, antibody-based therapies have been extensively used in HER2+ breast cancer. However, some patients inherently display resistance and in advanced stages, almost all eventually progress. Functionalized NPs through conjugation with antibodies appear to be a promising strategy to optimize targeted therapies due to properties related to biocompatibility, suitable delivery control and efficiency of functionalization. This review is focused on the different strategies to conjugate antibodies into polymeric NPs. Recent antibody conjugation approaches applied to the improvement of breast cancer therapy are highlighted in this review.
Collapse
Affiliation(s)
- Alberto Juan
- Oncología traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
| | - Francisco J. Cimas
- Oncología traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
| | - Iván Bravo
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer-CSIC, IBSAL- Salamanca and CIBERONC, 37007 Salamanca, Spain;
| | - Alberto Ocaña
- Oncología traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Experimental Therapeutics Unit, Hospital clínico San Carlos, IdISSC and CIBERONC, 28040 Madrid, Spain
- Correspondence: (A.O.); (C.A.-M.); Tel.: +34-635-681806 (A.O.); +34-9675-99200 (C.A.-M)
| | - Carlos Alonso-Moreno
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
- Correspondence: (A.O.); (C.A.-M.); Tel.: +34-635-681806 (A.O.); +34-9675-99200 (C.A.-M)
| |
Collapse
|
31
|
Pandya AD, Øverbye A, Sahariah P, Gaware VS, Høgset H, Masson M, Høgset A, Mælandsmo GM, Skotland T, Sandvig K, Iversen TG. Drug-Loaded Photosensitizer-Chitosan Nanoparticles for Combinatorial Chemo- and Photodynamic-Therapy of Cancer. Biomacromolecules 2020; 21:1489-1498. [PMID: 32092254 PMCID: PMC7307886 DOI: 10.1021/acs.biomac.0c00061] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
![]()
In
this study we have developed biodegradable polymeric nanoparticles
(NPs) containing the cytostatic drugs mertansine (MRT) or cabazitaxel
(CBZ). The NPs are based on chitosan (CS) conjugate polymers synthesized
with different amounts of the photosensitizer tetraphenylchlorin (TPC).
These TPC–CS NPs have high loading capacity and strong drug
retention due to π–π stacking interactions between
the drugs and the aromatic photosensitizer groups of the polymers.
CS polymers with 10% of the side chains containing TPC were found
to be optimal in terms of drug loading capacity and NP stability.
The TPC–CS NPs loaded with MRT or CBZ displayed higher cytotoxicity
than the free form of these drugs in the breast cancer cell lines
MDA-MB-231 and MDA-MB-468. Furthermore, light-induced photochemical
activation of the NPs elicited a strong photodynamic therapy effect
on these breast cancer cells. Biodistribution studies in mice showed
that most of the TPC–CS NPs accumulated in liver and lungs,
but they were also found to be localized in tumors derived from HCT-116
cells. These data suggest that the drug-loaded TPC–CS NPs have
a potential in combinatory anticancer therapy and as contrast agents.
Collapse
Affiliation(s)
- Abhilash D Pandya
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital,N-0379 Oslo, Norway
| | - Anders Øverbye
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, N-0379 Oslo, Norway
| | - Priyanka Sahariah
- Faculty of Pharmaceutical Sciences, School of Health Sciences, University of Iceland, Hofsvallagata 53, IS-107 Reykjavik, Iceland
| | - Vivek S Gaware
- Faculty of Pharmaceutical Sciences, School of Health Sciences, University of Iceland, Hofsvallagata 53, IS-107 Reykjavik, Iceland
| | - Håkon Høgset
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, N-0379 Oslo, Norway
| | - Màr Masson
- Faculty of Pharmaceutical Sciences, School of Health Sciences, University of Iceland, Hofsvallagata 53, IS-107 Reykjavik, Iceland
| | - Anders Høgset
- PCI Biotech AS, Ullernchause'en 64, N-0379 Oslo, Norway
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital,N-0379 Oslo, Norway.,Institute of Medical Biology, Faculty of Health Sciences, The Arctic University of Norway, University of Tromsø, Tromsø, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, N-0379 Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, N-0379 Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Tore-Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, N-0379 Oslo, Norway
| |
Collapse
|
32
|
Szwed M, Torgersen ML, Kumari RV, Yadava SK, Pust S, Iversen TG, Skotland T, Giri J, Sandvig K. Biological response and cytotoxicity induced by lipid nanocapsules. J Nanobiotechnology 2020; 18:5. [PMID: 31907052 PMCID: PMC6943936 DOI: 10.1186/s12951-019-0567-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022] Open
Abstract
Background Lipid nanocapsules (LNCs) are promising vehicles for drug delivery. However, since not much was known about cellular toxicity of these nanoparticles in themselves, we have here investigated the mechanisms involved in LNC-induced intoxication of the three breast cancer cell lines MCF-7, MDA-MD-231 and MDA-MB-468. The LNCs used were made of Labrafac™ Lipophile WL1349, Lipoid® S75 and Solutol® HS15. Results High resolution SIM microscopy showed that the DiD-labeled LNCs ended up in lysosomes close to the membrane. Empty LNCs, i.e. without encapsulated drug, induced not only increased lysosomal pH, but also acidification of the cytosol and a rapid inhibition of protein synthesis. The cytotoxicity of the LNCs were measured for up to 72 h of incubation using the MTT assay and ATP measurements in all three cell lines, and revealed that MDA-MB-468 was the most sensitive cell line and MCF-7 the least sensitive cell line to these LNCs. The LNCs induced generation of reactive free oxygen species and lipid peroxidation. Experiments with knock-down of kinases in the near-haploid cell line HAP1 indicated that the kinase HRI is essential for the observed phosphorylation of eIF2α. Nrf2 and ATF4 seem to play a protective role against the LNCs in MDA-MB-231 cells, as knock-down of these factors sensitizes the cells to the LNCs. This is in contrast to MCF-7 cells where the knock-down of these factors had a minor effect on the toxicity of the LNCs. Inhibitors of ferroptosis provided a large protection against LNC toxicity in MDA-MB-231 cells, but not in MCF-7 cells. Conclusions High doses of LNCs showed a different degree of toxicity on the three cell lines studied, i.e. MCF-7, MDA-MD-231 and MDA-MB-468 and affected signaling factors and the cell fate differently in these cell lines.
Collapse
Affiliation(s)
- Marzena Szwed
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Maria Lyngaas Torgersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Remya Valsala Kumari
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, India
| | - Sunil Kumar Yadava
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, India
| | - Sascha Pust
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Tore Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, India.
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway. .,Department of Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
33
|
Pandya AD, Jäger E, Bagheri Fam S, Höcherl A, Jäger A, Sincari V, Nyström B, Štěpánek P, Skotland T, Sandvig K, Hrubý M, Mælandsmo GM. Paclitaxel-loaded biodegradable ROS-sensitive nanoparticles for cancer therapy. Int J Nanomedicine 2019; 14:6269-6285. [PMID: 31496685 PMCID: PMC6689768 DOI: 10.2147/ijn.s208938] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/13/2019] [Indexed: 12/15/2022] Open
Abstract
Background Reactive oxygen species (ROS), such as hydrogen peroxide and superoxide, trigger biodegradation of polymer-based nanoparticles (NPs) bearing pinacol-type boronic ester groups. These NPs may selectively release their cargo, in this case paclitaxel (PTX), at the high levels of ROS present in the intracellular environment of inflamed tissues and most tumors. Purpose The main objective was to determine anti-tumor efficacy of PTX-loaded ROS-sensitive NPs and to examine whether macrophage infiltration had any impact on treatment efficacy. Methods NPs were synthesized and their characteristics in the presence of H2O2 were demonstrated. Both confocal microscopy as well as flow cytometry approaches were used to determine degradation of ROS-sensitive NPs. HeLa cells were cultured in vitro and used to establish tumor xenografts in nude mice. In vivo experiments were performed to understand toxicity, biodistribution and anti-tumor efficacy of the NPs. Moreover, we performed immunohistochemistry on tumor sections to study infiltration of M1 and M2 subsets of macrophages. Results We demonstrated that PTX delivered in NPs containing a ROS-sensitive polymer exhibits a better anti-tumor efficacy than PTX in NPs containing ROS-non-sensitive polymer, free PTX or Abraxane® (nab-PTX). The biodistribution revealed that ROS-sensitive NPs exhibit retention in liver, spleen and lungs, suggesting a potential to target cancer metastasizing to these organs. Finally, we demonstrated a correlation between infiltrated macrophage subsets and treatment efficacy, possibly contributing to the efficient anti-tumor effects. Conclusion Treatment with ROS-sensitive NPs containing PTX gave an improved therapeutic effect in HeLa xenografts than their counterpart, free PTX or nab-PTX. Our data revealed a correlation between macrophage infiltration and efficiency of the different antitumor treatments, as the most effective NPs resulted in the highest infiltration of the anti-tumorigenic M1 macrophages.
Collapse
Affiliation(s)
- Abhilash D Pandya
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Eliézer Jäger
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Shahla Bagheri Fam
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Anita Höcherl
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Alessandro Jäger
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Vladimir Sincari
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Bo Nyström
- Department of Chemistry, University of Oslo, Oslo, Norway
| | - Petr Štěpánek
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Martin Hrubý
- Institute of Macromolecular Chemistry v.v.i, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute of Medical Biology, Faculty of Health Sciences, The Arctic University of Norway - University of Tromsø, Tromsø, Norway
| |
Collapse
|
34
|
Vauthier C. A journey through the emergence of nanomedicines with poly(alkylcyanoacrylate) based nanoparticles. J Drug Target 2019; 27:502-524. [PMID: 30889991 DOI: 10.1080/1061186x.2019.1588280] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Starting in the late 1970s, the pioneering work of Patrick Couvreur gave birth to the first biodegradable nanoparticles composed of a biodegradable synthetic polymer. These nanoparticles, made of poly(alkylcyanoacrylate) (PACA), were the first synthetic polymer-based nanoparticulate drug carriers undergoing a phase III clinical trial so far. Analyzing the journey from the birth of PACA nanoparticles to their clinical evaluation, this paper highlights their remarkable adaptability to bypass various drug delivery challenges found on the way. At present, PACA nanoparticles include a wide range of nanoparticles that can associate drugs of different chemical nature and can be administered in vivo by different routes. The most recent technologies giving the nanoparticles customised functions could also be implemented on this family of nanoparticles. Through different examples, this paper discusses the seminal role of the PACA nanoparticles' family in the development of nanomedicines.
Collapse
Affiliation(s)
- Christine Vauthier
- a Institut Galien Paris Sud, UMR CNRS 8612 , Université Paris-Sud , Chatenay-Malabry Cedex , France
| |
Collapse
|