1
|
Dykman L, Khlebtsov B, Khlebtsov N. Drug delivery using gold nanoparticles. Adv Drug Deliv Rev 2025; 216:115481. [PMID: 39617254 DOI: 10.1016/j.addr.2024.115481] [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: 07/12/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
Modern nanotechnologies provide various possibilities for efficiently delivering drugs to biological targets. This review focuses on using functionalized gold nanoparticles (GNPs) as a drug delivery platform. Owing to their exceptional size and surface characteristics, GNPs are a perfect drug delivery vehicle for targeted and selective distribution. Several in vitro and in vivo tests have shown how simple it is to tailor these particles to administer chemical medications straight to tumors. The GNP surface can also be coated with ligands to modify drug release or improve selectivity. Moreover, the pharmacological activity can be enhanced by using the photothermal characteristics of the particles.
Collapse
Affiliation(s)
- Lev Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Boris Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Nikolai Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia; Saratov State University, 83 Ulitsa Astrakhanskaya, Saratov 410012, Russia.
| |
Collapse
|
2
|
Salehi S, Boddohi S, Adel Ghiass M, Behmanesh M. Microfluidic preparation and optimization of (Kollicoat ® IR-b-PCL) polymersome for co-delivery of Nisin-Curcumin in breast cancer application. Int J Pharm 2024; 660:124371. [PMID: 38908809 DOI: 10.1016/j.ijpharm.2024.124371] [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: 04/05/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
This work aimed to develop amphiphilic nanocarriers such as polymersome based diblock copolymer of Kollicoat ® IR -block-poly(ε-caprolactone) (Kollicoat ® IR-b-PCL) for potential co-delivery of Nisin (Ni) and Curcumin (CUR) for treatment of breast cancer. To generate multi-layered nanocarriers of uniform size and morphology, microfluidics was used as a new technology. In order to characterise and optimize polymersome, design of experiments (Design-Expert) software with three levels full factorial design (3-FFD) method was used. Finally, the optimized polymersome was produced with a spherical morphology, small particle size (dH < 200 nm), uniform size distribution (PDI < 0.2), and high drug loading efficiency (Ni 78 % and CUR 93 %). Furthermore, the maximum release of Ni and CUR was found to be roughly 60 % and 80 % in PBS, respectively. Cytotoxicity assays showed a slight cytotoxicity of Ni and CUR -loaded polymersome (N- Ni /CUR) towards normal cells while demonstrating inhibitory activity against cancer cells compared to the free drugs. Also, the apoptosis assays and cellular uptake confirmed the obtained results from cytotoxic analysis. In general, this study demonstrated a microfluidic approach for preparation and optimization of polymersome for co-delivery of two drugs into cancer cells.
Collapse
Affiliation(s)
- Sahar Salehi
- Department of Biomedical Engineering, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Soheil Boddohi
- Department of Biomedical Engineering, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran.
| | - Mohammad Adel Ghiass
- Tissue Engineering Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mehrdad Behmanesh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
3
|
Dhull A, Wei J, Pulukuri AJ, Rani A, Sharma R, Mesbahi N, Yoon H, Savoy EA, Xaivong Vi S, Goody KJ, Berkman CE, Wu BJ, Sharma A. PSMA-targeted dendrimer as an efficient anticancer drug delivery vehicle for prostate cancer. NANOSCALE 2024; 16:5634-5652. [PMID: 38440933 DOI: 10.1039/d3nr06520k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related deaths among men in the United States. Although early-stage treatments exhibit promising 5-year survival rates, the treatment options for advanced stage disease are constrained, with short survival benefits due to the challenges associated with effective and selective drug delivery to PCa cells. Even though targeting Prostate Specific Membrane Antigen (PSMA) has been extensively explored and is clinically employed for imaging and radio-ligand therapy, the clinical success of PSMA-based approaches for targeted delivery of chemotherapies remains elusive. In this study, we combine a generation 4 hydroxy polyamidoamine dendrimer (PD) with irreversible PSMA ligand (CTT1298) to develop a PSMA-targeted nanoplatform (PD-CTT1298) for selective intracellular delivery of potent chemotherapeutics to PCa. PD-CTT1298-Cy5 exhibits a PSMA IC50 in the nanomolar range and demonstrates selective uptake in PSMA (+) PCa cells via PSMA mediated internalization. When systemically administered in a prostate tumor xenograft mouse model, PD-CTT1298-Cy5 selectively targets PSMA (+) tumors with significantly less accumulation in PSMA (-) tumors or upon blocking of the PSMA receptors. Moreover, the dendrimer clears rapidly from the off-target organs limiting systemic side-effects. Further, the conjugation of an anti-cancer agent, cabozantinib to the PSMA-targeted dendrimer translates to a significantly enhanced anti-proliferative activity in vitro compared to the free drug. These findings highlight the potential of PD-CTT1298 nanoplatform as a versatile approach for selective delivery of high payloads of potent chemotherapeutics to PCa, where dose related systemic side-effects are a major concern.
Collapse
Affiliation(s)
- Anubhav Dhull
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Jing Wei
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Anunay James Pulukuri
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Anu Rani
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Rishi Sharma
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Nooshin Mesbahi
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Hosog Yoon
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Emily A Savoy
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Sylvia Xaivong Vi
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Kenneth John Goody
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Clifford E Berkman
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Anjali Sharma
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| |
Collapse
|
4
|
Saddam Hussain M, Khetan R, Clulow AJ, Ganesan R, MacMillan A, Robinson N, Ahmed-Cox A, Krasowska M, Albrecht H, Blencowe A. Teaching an Old Dog New Tricks: A Global Approach to Enhancing the Cytotoxicity of Drug-Loaded, Non-responsive Micelles Using Oligoelectrolytes. ACS APPLIED MATERIALS & INTERFACES 2024; 16:9736-9748. [PMID: 38349780 DOI: 10.1021/acsami.3c16551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Polymeric micelles have been extensively studied as vectors for the delivery of hydrophobic drugs for the treatment of cancers and other diseases. Despite intensive research, few formulations provide significant benefits, and even fewer have been clinically approved. While many traditional non-responsive micelles have excellent safety profiles, they lack the ability to respond to the intracellular environment and release their cargo in a spatiotemporally defined manner to effectively deliver large doses of cytotoxic drugs into the cytosol of cells that overwhelm efflux pumps. As a novel and adaptable strategy, we hypothesized that well-established non-responsive polymeric micelles could be augmented with a pH-trigger via the co-encapsulation of cytocompatible oligoelectrolytes, which would allow rapid cargo release in the endosome, leading to increased cytotoxicity. Herein, we demonstrate how this strategy can be applied to render non-responsive micelles pH-responsive, resulting in abrupt cargo release at specific and tunable pH values compatible with endosomal delivery, which significantly increased their cytotoxicity up to 3-fold in an ovarian adenocarcinoma (SKOV-3) cell line compared to non-responsive micelles. In comparison, the oligoelectrolyte-loaded micelles were significantly less toxic to healthy 3T3 fibroblasts, indicating a selective cargo release in cancer cell lines. Oligoelectrolytes can be co-encapsulated in the micelles along with drugs at high encapsulation efficiency percentages, which are both ejected from the micelle core upon oligoelectrolyte ionization. Mechanistically, the increase in cytotoxicity appears to also result from the accelerated endosomal escape of the cargo caused by disruption of the endosomal membrane by the simultaneous release of the oligoelectrolytes from the micelles. Furthermore, we show how this approach is broadly applicable to non-responsive micelles regardless of their composition and various classes of hydrophobic chemotherapeutics. The preliminary studies presented here reveal the versatility and wide scope of oligoelectrolyte-mediated, pH-triggered drug release as a compelling and powerful strategy to enhance the cytotoxicity of non-responsive polymeric micelles.
Collapse
Affiliation(s)
- Md Saddam Hussain
- Applied Chemistry and Translational Biomaterials (ACTB) Group, Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Riya Khetan
- Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Andrew J Clulow
- Australian Synchrotron, Australian Nuclear Science and Technology Organisation (ANSTO), 800 Blackburn Road, Clayton, Victoria 3168, Australia
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Raja Ganesan
- Centre for Cancer Biology, UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Alexander MacMillan
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, New South Wales 2033, Australia
| | - Nirmal Robinson
- Centre for Cancer Biology, UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Aria Ahmed-Cox
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, New South Wales 2033, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales 2750, Australia
- Australian Centre for Nanomedicine, Faculty of Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Marta Krasowska
- Surface Interactions and Soft Matter (SISM) Group, Future Industries Institute, UniSA STEM, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Hugo Albrecht
- Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Anton Blencowe
- Applied Chemistry and Translational Biomaterials (ACTB) Group, Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| |
Collapse
|
5
|
Hasan U, Rajakumara E, Giri J. Reversal of Multidrug Resistance by the Synergistic Effect of Reversan and Hyperthermia to Potentiate the Chemotherapeutic Response of Doxorubicin in Glioblastoma and Glioblastoma Stem Cells. ACS APPLIED BIO MATERIALS 2023; 6:5399-5413. [PMID: 37975516 DOI: 10.1021/acsabm.3c00644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
The glioblastoma stem cell (GSC) population in glioblastoma multiforme (GBM) poses major complication in clinical oncology owing to increased resistance to chemotherapeutic drugs, thereby limiting treatment in patients with recurring glioblastoma. To completely eradicate glioblastoma, a single therapy module is not enough; therefore, there is a need to develop a multimodal approach to eliminate bulk tumors along with the CSC population. With an aim to target transporters associated with multidrug resistance (MDR), such as P-glycoprotein (P-gp), a small-molecule inhibitor, reversan (RV) was used along with multifunctional magnetic nanoparticles (MNPs) for hyperthermia (HT) therapy and targeted drug delivery. Higher efflux of free doxorubicin (Dox) from the cells was stabilized by encapsulation in PPS-MnFe nanoparticles, whose physicochemical properties were determined by dynamic light scattering (DLS) and transmission electron microscopy (TEM). Treatment with RV also enhanced the cellular uptake of PPS-MnFe-Dox, whereas RV and magnetic hyperthermia (MHT) together showed prolonged retention of fluorescence dye, Rhodamine123 (R123), in glioblastoma cells compared with individual treatment. Overall, in this work, we demonstrated the synergistic action of RV and HT to combat MDR in GBM and GSCs, and chemo-hyperthermia therapy enhanced the cytotoxic effect of the chemotherapeutic drug Dox (with lower effective concentration) and induced a higher degree of apoptosis compared to single-drug dosage.
Collapse
Affiliation(s)
- Uzma Hasan
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502284, India
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502284, India
| | - Eerappa Rajakumara
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502284, India
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502284, India
| |
Collapse
|
6
|
Sultana N, Ali A, Waheed A, Jabi B, Yaqub Khan M, Mujeeb M, Sultana Y, Aqil M. Dissolving microneedle transdermal patch loaded with Risedronate sodium and Ursolic acid bipartite nanotransfersomes to combat osteoporosis: Optimization, characterization, in vitro and ex vivo assessment. Int J Pharm 2023; 644:123335. [PMID: 37597597 DOI: 10.1016/j.ijpharm.2023.123335] [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: 03/28/2023] [Revised: 07/30/2023] [Accepted: 08/17/2023] [Indexed: 08/21/2023]
Abstract
Osteoporosis is a fatal bone-wearing malady and a substantial reason behind the impermanence of human life and economic burden. Risedronate Sodium along with Ursolic acid has been studied to ameliorate osteoporosis. To bypass problems associated with bioavailability, we have developed a microneedle transdermal patch loaded with optimized formulation nanotransfersomes. It was optimized using three factor, three-level Central composite design with independent variables namely, the concentration of phospholipid, surfactant, and sonication time on dependent variables (vesicle size, entrapment efficiency and Polydispersity index). Vesicles of size 271.9 ± 8.45 nm with PDI 0.184 ± 0.01, having entrapment efficiency of 86.12 ± 5.20% and 85.65 ± 4.88% for RIS and UA respectively were observed. In vitro release study showed the sustained release pattern with 78.16 ± 1.12% and 75.72 ± 1.01% release of RIS and UA respectively. Dissolving MN patch prepared from gelatin was found to have good strength and folding endurance with uniform drug content (98.68 ± 0.004%). Ex vivo permeation study revealed that up to 80% of the drug can be permeated within 24 h. CLSM analysis was also performed to show penetration of RU-NTRs. From the results obtained, we can conclude that dissolving MN patch loaded with RU-NTRs has great potential than its conventional counterpart.
Collapse
Affiliation(s)
- Niha Sultana
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Asad Ali
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Ayesha Waheed
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Bushra Jabi
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Yaqub Khan
- Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan City 320314, Taiwan
| | - Mohd Mujeeb
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Yasmin Sultana
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Aqil
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
7
|
Yu HR, Chen BH. Analysis of Phenolic Acids and Flavonoids in Rabbiteye Blueberry Leaves by UPLC-MS/MS and Preparation of Nanoemulsions and Extracts for Improving Antiaging Effects in Mice. Foods 2023; 12:1942. [PMID: 37238760 PMCID: PMC10216987 DOI: 10.3390/foods12101942] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/02/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Rabbiteye blueberry leaves, a waste produced after harvest of blueberry, are rich in polyphenols. This study aims to analyze phenolic acids and flavonoids in blueberry leaves by UPLC-MS/MS and prepare nanoemulsions for determining anti-aging activity in mice. Overall, 30% ethanol was the most suitable extraction solvent for total phenolic acids and total flavonoids. A total of four phenolic acids and four flavonoids were separated within seven minutes for further identification and quantitation by UPLC-MS/MS in selective reaction monitoring (SRM) mode, with 3-O-caffeoylquinic acid being present in the highest amount (6474.2 μg/g), followed by quercetin-3-O-galactoside (1943.9 μg/g), quercetin-3-O-rutinoside (1036.6 μg/g), quercetin-3-O-glucoside (867.2 μg/g), 5-O-caffeoylquinic acid (815.8 μg/g), kaempferol-3-O-glucoside (309.7 μg/g), 3,5-dicaffeoylquinic acid (195.3 μg/g), and 4,5-dicaffeoylquinic acid (60.8 μg/g). The blueberry nanoemulsion was prepared by using an appropriate ratio of soybean oil, Tween 80, glycerol, ethanol, and water at 1.2%, 8%, 2%, 2%, and 86.8%, respectively, and mixing with dried blueberry extract, with the mean particle size and zeta potential being 16 nm and -54 mV, respectively. A high stability was observed during storage of nanoemulsion for 90 days at 4 °C and heated at 100 °C for 2 h. An animal study revealed that this nanoemulsion could elevate dopamine content in mice brain as well as superoxide dismutase, glutathione peroxidase, and catalase activities in mice liver while reducing the contents of malondialdehyde and protein carbonyl in mice brains. Collectively, the high-dose nanoemulsion possessed the highest efficiency in improving mice aging with a promising potential for development into a health food.
Collapse
Affiliation(s)
- Hsin-Rong Yu
- Department of Food Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
| | - Bing-Huei Chen
- Department of Food Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
- Department of Nutrition, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
8
|
Simon AT, Chattopadhyay A, Ghosh SS. In Vitro Therapeutic Attributes of Luminescent Hydroxyapatite Nanoparticles in Codelivery Module. ACS APPLIED BIO MATERIALS 2022; 5:2741-2753. [PMID: 35608933 DOI: 10.1021/acsabm.2c00201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Imminent prospects of clinical importance have been accomplished through divergent treatment modalities implemented using nanoscale platforms. In the present study, hydroxyapatite nanoparticles doped with copper nanoclusters (HAPs) were explored for codelivery of a hydrophobic drug, namely, norfloxacin (NX), and a hydrophilic photosensitizer, such as methylene blue (MB). NX and MB were successfully homed into HAPs (MB-NX-HAPs), which further exhibited a pH-dependent release of both. With the objective of attaining an enhanced effect, MB-NX-HAPs were evaluated for combination therapy, involving chemotherapy and photodynamic therapy (PDT) with irradiation at 640 nm. The combinatorial therapy approach was initially applied for antibacterial therapy, which suggested a considerable reduction in bacterial growth of Gram-negative strain Pseudomonas aeruginosa MTCC 2488. Thereafter, the antiproliferative study performed in cancer cell lines (HeLa and MCF-7) revealed the efficiency of MB-NX-HAPs in bestowing a combinatorial effect through chemotherapy and PDT (irradiation at 640 nm). The combined effect exerted through MB-NX-HAPs subsequently induced reactive oxygen species (ROS) generation, cell cycle alteration, and apoptosis activation in cancer cells. The biocompatible nature of MB-NX-HAPs was appreciably shown through their minimal effect on the normal cell line (HEK-293). Additionally, HAPs through luminescence of copper nanoclusters were suggested to aid in bioimaging of cancer cell lines.
Collapse
Affiliation(s)
- Anitha T Simon
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Arun Chattopadhyay
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, India.,Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati781039, India
| | - Siddhartha Sankar Ghosh
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, India.,Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati781039, India
| |
Collapse
|
9
|
Liao TT, Han JF, Zhang FY, Na R, Ye WL. Enhanced Anti-Tumor Effect of Folate-Targeted FA-AMA-hyd-DOX Conjugate in a Xenograft Model of Human Breast Cancer. Molecules 2021; 26:molecules26237110. [PMID: 34885691 PMCID: PMC8659115 DOI: 10.3390/molecules26237110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/19/2021] [Accepted: 11/19/2021] [Indexed: 11/16/2022] Open
Abstract
Folate-aminocaproic acid-doxorubicin (FA-AMA-hyd-DOX) was firstly synthesized by our group. It was indicated that FA-AMA-hyd-DOX was pH-responsive, and had strong cytotoxicity on a folate receptor overexpressing cell line (KB cells) in vitro. The aim of our study was to further explore the potential use of FA-AMA-hyd-DOX as a new therapeutic drug for breast cancer. The cellular uptake and the antiproliferative activity of the FA-AMA-hyd-DOX in MDA-MB-231 cells were measured. Compared with DOX, FA-AMA-hyd-DOX exhibited higher targeting ability and cytotoxicity to FR-positive tumor cells. Subsequently, the tissue distribution of FA-AMA-hyd-DOX was studied, and the result confirmed that DOX modified by FA can effectively increase the selectivity of drugs in vivo. After determining the maximum tolerated dose (MTD) of FA-AMA-hyd-DOX in MDA-MB-231 tumor-bearing nude mice, the antitumor effects and the in vivo safety of FA-AMA-hyd-DOX were systematically evaluated. The data showed that FA-AMA-hyd-DOX could effectively increase the dose of DOX tolerated by tumor-bearing nude mice and significantly inhibit MDA-MB-231 tumor growth in vivo. Furthermore, FA-AMA-hyd-DOX treatment resulted in almost no obvious damage to the mice. All the positive data suggest that FA-targeted FA-AMA-hyd-DOX is a promising tumor-targeted compound for breast cancer therapy.
Collapse
Affiliation(s)
- Tian-tian Liao
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an 710032, China; (T.-t.L.); (F.-y.Z.)
| | - Jiang-fan Han
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Fei-yue Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an 710032, China; (T.-t.L.); (F.-y.Z.)
| | - Ren Na
- Department of Epidemiology and Health Statistics, Faculty of Military Preventive Medicine, Fourth Military Medical University, Xi’an 710032, China;
| | - Wei-liang Ye
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an 710032, China; (T.-t.L.); (F.-y.Z.)
- Correspondence: ; Tel.: +86-29-8477-6783
| |
Collapse
|
10
|
Gyanewali S, Kesharwani P, Sheikh A, Ahmad FJ, Trivedi R, Talegaonkar S. Formulation development and in vitro-in vivo assessment of protransfersomal gel of anti-resorptive drug in osteoporosis treatment. Int J Pharm 2021; 608:121060. [PMID: 34500057 DOI: 10.1016/j.ijpharm.2021.121060] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022]
Abstract
Osteoporosis is a major cause of morbidity, mortality, and economic burden worldwide. Despite being an effective in combating the bone-deteriorating disorders, bisphosphonates have several shortcomings including poor and variable bioavailability, low permeability, high toxicity, etc. In this study, we developed and optimized protransfersome formulation for the drug risedronate sodium (RIS-Na) with the goal of enhancing its bioavailability and hence patient compliance. Phase separation coacervation technique was utilized for development of optimized formulation. Optimization was achieved by using three-factor, three-level Box-Behnken design combined with Response Surface Methodology (RSM). This enabled us to decipher the effect of 3 independent variables (Phospholipid, Tween-80 and Sodium Deoxycholate) on three dependent parameters (entrapment efficiency, vesicle size and transdermal flux). Optimized formulation was further evaluated for pharmacokinetic and pharmacodynamic parameters. Smooth, spherical protransfersomes with a size of 260 ± 18 nm, having entrapment efficiency and flux of 80.4 ± 4.90% and 8.41 ± 0.148 μg/cm2/h, respectively were prepared. Ex vivo studies revealed a shorter lag time of 1.21 ± 0.18 h and higher flux associated with transdermal formulation. CLSM analysis further revealed better drug penetration (220 μm) through the skin in case of protransfersomes as compared to drug solution (72 μm). Additionally, biomechanical, biochemical, and histo-pathological studies further validated the results. Thus, it was concluded that protransfersome formulation has a great potential in providing better therapeutic efficacy of risedronate than its conventional counterpart.
Collapse
Affiliation(s)
- Suman Gyanewali
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India.
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Ritu Trivedi
- Department of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sushama Talegaonkar
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India.
| |
Collapse
|
11
|
Curcumin-Loaded Iron Oxide Nanoparticles Coated with Sodium Alginate and Hydroxyapatite and Their Cytotoxic Effects Against the HT-29 and MCF-7 Cancer Cell Lines. Jundishapur J Nat Pharm Prod 2021. [DOI: 10.5812/jjnpp.114572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Curcumin, a bioactive component of Curcuma langa, has been investigated for its anti-proliferative effects against various cancer cell lines. Although results are very promising, the poor water solubility and low bioavailability of curcumin are its main limitations for clinical application. Objectives: The purpose of this study was to develop a drug delivery system, consisting of hydroxyapatite (HAp) polymer and sodium alginate (NaAlg), covering the magnetic core of iron oxide nanoparticles (IONPs), and loaded with curcumin in order to enhance its bioavailability and therapeutic efficacy. Methods: In this study, IONPs were prepared by the co-precipitation method and coated with HAp and NaAlg. The nanoparticles (NPs) were characterized by X-ray diffraction, Fourier Transform Infrared Spectroscopy (FTIR), and electron microscopy (TEM and SEM). Encapsulation efficiency and curcumin loading rate were examined. Drug release rate was also measured in vitro at pH = 7.5 and 5.5. The toxicity of curcumin-loaded NPs and free curcumin was evaluated against HT-29 and MCF-7 cancer cells. Results: The assessment of physicochemical characteristics showed the synthesis of spherical particles with nanometer sizes (5 - 7 nm) and a high encapsulation efficiency (84.16 ± 3.51 %) and drug loading capacity (21.03 ± 0.87%). Maximum drug release was obtained at pH = 5.5. Iron oxide nanoparticles showed no significant cytotoxic effects. Curcumin-loaded coated IONPs showed a higher toxicity against HT-29 and MCF-7 cancer cells compared to free curcumin. Conclusions: This in vitro study showed that the encapsulation of curcumin, as a potent herbal drug, into IONPs enhanced its bioavailability, suggesting the NPs as an efficient vehicle for targeted drug delivery in cancer treatment.
Collapse
|
12
|
Alhowail AH, Pinky PD, Eggert M, Bloemer J, Woodie LN, Buabeid MA, Bhattacharya S, Jasper SL, Bhattacharya D, Dhanasekaran M, Escobar M, Arnold RD, Suppiramaniam V. Doxorubicin induces dysregulation of AMPA receptor and impairs hippocampal synaptic plasticity leading to learning and memory deficits. Heliyon 2021; 7:e07456. [PMID: 34296005 PMCID: PMC8282984 DOI: 10.1016/j.heliyon.2021.e07456] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/10/2021] [Accepted: 06/28/2021] [Indexed: 11/27/2022] Open
Abstract
Doxorubicin (Dox) is a chemotherapeutic agent used widely to treat a variety of malignant cancers. However, Dox chemotherapy is associated with several adverse effects, including "chemobrain," the observation that cancer patients exhibit through learning and memory difficulties extending even beyond treatment. This study investigated the effect of Dox treatment on learning and memory as well as hippocampal synaptic plasticity. Dox-treated mice (5 mg/kg weekly x 5) demonstrated impaired performance in the Y-maze spatial memory task and a significant reduction in hippocampal long-term potentiation. The deficit in synaptic plasticity was mirrored by deficits in the functionality of synaptic `α-amino-3- hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) channels, including reduced probability of opening, decreased dwell open time, and increased closed times. Furthermore, a reduction in the AMPAR subunit GluA1 level, its downstream signaling molecule Ca2+/calmodulin-dependent protein kinase (CaMKII), and brain-derived neurotrophic factor (BDNF) were observed. This was also accompanied by an increase in extracellular signal regulated kinase (ERK) and protein kinase B (AKT) activation. Together these data suggest that Dox-induced cognitive impairments are at least partially due to alterations in the expression and functionality of the glutamatergic AMPAR system.
Collapse
Affiliation(s)
- Ahmad H. Alhowail
- Department of Pharmacology and Toxicology, Qassim University, Buraydah, Saudi Arabia
| | - Priyanka D. Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama, USA
| | - Matthew Eggert
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama, USA
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama, USA
- Department of Pharmaceutical and Biomedical Sciences, Touro College of Pharmacy, New York, NY, USA
| | - Lauren N. Woodie
- Department of Nutrition, Dietetics and Hospitality Management, College of Human Sciences, Auburn University, Auburn, Alabama, USA
- Institute for Diabetes, Obesity and Metabolism, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Manal A. Buabeid
- College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| | - Shanese L. Jasper
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama, USA
| | | | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| | - Martha Escobar
- Department of Psychology, Oakland University, Rochester, MI, USA
| | - Robert D. Arnold
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| |
Collapse
|
13
|
Guncum E, Bakirel T, Anlas C, Isiklan N, Ustun Alkan F, Charehsaz M, Aydin A. The screening of the safety profile of polymeric based amoxicillin nanoparticles in various test systems. Toxicol Lett 2021; 348:1-9. [PMID: 33984417 DOI: 10.1016/j.toxlet.2021.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/18/2021] [Accepted: 05/07/2021] [Indexed: 11/25/2022]
Abstract
Nanotechnology-based drugs show superiority over conventional medicines because of increased bioavailability, lower accumulation in non-target tissues, and improved therapeutic index with increased accumulation at target sites. However, it is important to be aware of possible problems related to the toxicity of these products, which have therapeutically superior properties. Accordingly, the present study was designed to investigate the safety profile of amoxicillin nanoparticles (AmxNPs) that we developed to increase the oral bioavailability of amoxicillin (Amx) in poultry. In the first part of the study, the genotoxicity potential of AmxNPs was evaluated using the Ames test and the in vitro comet assay. The results of Ames test showed that none of the tested concentrations of Amx and AmxNPs cause a significant increase in the revertant number of Salmonella typhimurium strains TA98, and TA100, either with or without metabolic activation. Similarly, the comet assay revealed that AmxNPs did not induce DNA damage at any of the concentrations used, whereas high-dose (200 μg/mL) of Amx caused a significant increase in the percentage of DNA in the tail. In the second part of the study, the toxicity potential of AmxNPs on broilers was investigated by measuring biochemical parameters. In vivo results demonstrated that AmxNps did not cause a significant change in biochemical parameters, whereas Amx increased ALT, glucose, and cholesterol levels at certain sampling times. The obtained findings suggest that AmxNPs could be a safe promising potential drug in drug delivery systems.
Collapse
Affiliation(s)
- Enes Guncum
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Kırıkkale University, Kırıkkale, Turkey.
| | - Tulay Bakirel
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Istanbul University- Cerrahpasa, Turkey.
| | - Ceren Anlas
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Istanbul University- Cerrahpasa, Turkey.
| | - Nuran Isiklan
- Department of Chemistry, Faculty of Science and Arts, Kırıkkale University, Kırıkkale, Turkey.
| | - Fulya Ustun Alkan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Istanbul University- Cerrahpasa, Turkey.
| | - Mohammad Charehsaz
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey.
| | - Ahmet Aydin
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
14
|
Schoppa T, Jung D, Rust T, Mulac D, Kuckling D, Langer K. Light-responsive polymeric nanoparticles based on a novel nitropiperonal based polyester as drug delivery systems for photosensitizers in PDT. Int J Pharm 2021; 597:120326. [PMID: 33540003 DOI: 10.1016/j.ijpharm.2021.120326] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/18/2022]
Abstract
Although nanoparticles (NPs) bear a great potential in tumour therapy, just a few nanosized drug delivery systems are commercially available. Besides their advantages like passive drug targeting and stable embedment of lipophilic active pharmaceutical ingredients, targeted drug release is a major challenge for a safe therapy. While drug release of commonly used materials depends on physiological factors, nanoparticles prepared by using stimuli responsive polymers offer a promising approach. External irradiation of light-sensitive nanoparticles enables local drug release, resulting in selective accumulation and consequently more effective treatment with less side effects. In this study light-responsive nanoparticles based on a new innovative light-responsive polyester (Nip-SLrPE) combined with poly(DL-lactide-co-glycolide) (PLGA) were prepared and examined for their physicochemical characteristics and light-triggered properties. As model drug the photosensitizer 5,10,15,20-tetrakis(m-hydroxyphenyl)chlorine (mTHPC) was incorporated and light-depending drug release was investigated. Furthermore, cytotoxic potential of selected formulations for PDT and intracellular accumulation of mTHPC were evaluated. In conclusion, nanoparticles based on the new light-sensitive Nip-SLrPE showed auspicious light-responsive properties, resulting in promising results for a smart drug delivery system.
Collapse
Affiliation(s)
- Timo Schoppa
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstraße 48, 48149 Muenster, Germany.
| | - Dimitri Jung
- Department of Chemistry, Paderborn University, Warburger Str. 100, 33098 Paderborn, Germany.
| | - Tarik Rust
- Department of Chemistry, Paderborn University, Warburger Str. 100, 33098 Paderborn, Germany.
| | - Dennis Mulac
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstraße 48, 48149 Muenster, Germany.
| | - Dirk Kuckling
- Department of Chemistry, Paderborn University, Warburger Str. 100, 33098 Paderborn, Germany.
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstraße 48, 48149 Muenster, Germany.
| |
Collapse
|
15
|
Li D, Xu L, Wang J, Gautrot JE. Responsive Polymer Brush Design and Emerging Applications for Nanotheranostics. Adv Healthc Mater 2021; 10:e2000953. [PMID: 32893474 PMCID: PMC11468394 DOI: 10.1002/adhm.202000953] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/11/2020] [Indexed: 12/29/2022]
Abstract
Responsive polymer brushes are a category of polymer brushes that are capable of conformational and chemical changes in response to external stimuli. They offer unique opportunities for the control of bio-nano interactions due to the precise control of chemical and structural parameters such as the brush thickness, density, chemistry, and architecture. The design of responsive brushes at the surface of nanomaterials for theranostic applications has developed rapidly. These coatings can be generated from a very broad range of nanomaterials, without compromising their physical, photophysical, and imaging properties. Although the use of responsive brushes for nanotheranostic remains in its early stages, in this review, the aim is to present how the systems developed to date can be combined to control sensing, imaging, and controlled delivery of therapeutics. The recent developments for such design and associated methods for the synthesis of responsive brushes are discussed. The responsive behaviors of homo polymer brushes and brushes with more complex architectures are briefly reviewed, before the applications of responsive brushes as smart delivery systems are discussed. Finally, the recent work is summarized on the use of responsive polymer brushes as novel biosensors and diagnostic tools for the detection of analytes and biomarkers.
Collapse
Affiliation(s)
- Danyang Li
- School of Cancer and Pharmaceutical SciencesKing's College London150 Stamford StreetLondonSE1 9NHUK
- Institute of BioengineeringQueen MaryUniversity of LondonMile End RoadLondonE1 4NSUK
- School of Engineering and Materials ScienceQueen MaryUniversity of LondonMile End RoadLondonE1 4NSUK
| | - Lizhou Xu
- Department of MaterialsImperial College LondonLondonSW7 2AZUK
| | - Jing Wang
- School of Life SciencesNorthwestern Polytechnical UniversityXi'an710072China
| | - Julien E. Gautrot
- Institute of BioengineeringQueen MaryUniversity of LondonMile End RoadLondonE1 4NSUK
- School of Engineering and Materials ScienceQueen MaryUniversity of LondonMile End RoadLondonE1 4NSUK
| |
Collapse
|
16
|
Phung CD, Tran TH, Choi JY, Jeong JH, Ku SK, Yong CS, Kim JO. Pre- and Post-Transcriptional Regulation of cFLIP for Effective Cancer Therapy Using pH-Ultrasensitive Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2021; 13:5999-6010. [PMID: 33506682 DOI: 10.1021/acsami.0c20624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cellular FLIP (cFLIP) is a crucial player of apoptosis-regulated pathways that is frequently overexpressed in solid cancers. To inhibit c-FLIP, pre- and post-transcriptionally, a multifunctional nanoparticle (NP) was created to deliver cFLIP-specific small interfering RNA (siRNA) into cancer cells. Specifically, Vorinostat (Vor)-loaded mesoporous silica nanoparticles (MSN) were conjugated with polyethylenimine-biotin (PB), followed by electrostatically binding with cFLIP siRNA (Vor/siR@MSN-PB). To stabilize and prolong the circulation time of nanoparticles, a bialdehyde-modified poly(ethylene glycol) (PEG) was cross-linked onto the polyethylenimine (PEI) backbone via the formation of the imine linkage (Schiff base) (Vor/siR@MSN-PB-PEG). The Schiff base is highly stable at physiological pH 7.4 but labile under slightly acidic pH conditions. In the acidic tumor microenvironment (TME), the PEG outer layer could be rapidly cleaved, resulting in the switching of the nanoparticle surface charge to positive, which specifically enhances internalization of the NPs to the biotin-positive tumor cells. Our results demonstrated the successful preparation of Vor/siR@MSN-PB-PEG NPs, in which the siRNA was effectively protected in serum and regulated the expression of cFlip, post-transcriptionally. The presence of the PEG layer resulted in high tumor accumulation and high efficacy in tumor inhibition, which was a result of the efficient cFLIP suppression. Furthermore, in the low-dose regimen of Vorinostat-the pre-transcriptional cFLIP suppressor, treatment with Vor/siR@MSN-PB-PEG NPs was found to be safe with the treated mice, indicating a promising combination regimen for cancer therapy.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Vietnam
- PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No. 167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Vietnam
| | - Ju-Yeon Choi
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
17
|
Kolluru LP, Chandran T, Shastri PN, Rizvi SAA, D’Souza MJ. Development and evaluation of polycaprolactone based docetaxel nanoparticle formulation for targeted breast cancer therapy. JOURNAL OF NANOPARTICLE RESEARCH 2020; 22:372. [DOI: 10.1007/s11051-020-05096-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/24/2020] [Indexed: 10/16/2023]
|
18
|
Cheah JUJ, Low HB, Zhang Y, Yong Kah JC. Light-independent M1 macrophage polarization by photosensitizer-loaded protein corona on gold nanorods. Nanomedicine (Lond) 2020; 15:2329-2344. [PMID: 32945247 DOI: 10.2217/nnm-2020-0249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To establish a light-independent functionality of gold nanorods (AuNRs) with a human serum (HS) protein corona loaded with photosensitizer Chlorin e6 (AuNR-HS-Ce6) in M1 polarization of macrophages. Methods: RT-qPCR and ELISA were used to determine gene and protein expression, respectively. Uptake of AuNR-HS-Ce6 was determined via flow cytometry, inductively coupled plasma mass spectrometry and fluorescence microscopy. Cell viability was determined using PrestoBlue® cell viability assay. Results: An increase in M1 gene and protein expression was observed in AuNR-HS-Ce6-treated macrophages. Delivery of high Ce6 payload via AuNR-HS-Ce6 was the primary contributor toward M1 polarization. Finally, DLD-1 cells treated with conditioned media from AuNR-HS-Ce6-treated macrophages showed significantly reduced proliferation. Conclusion: Our study suggests an immunomodulatory potential of Ce6 in inducing light-independent M1 polarization outside of its role as a photosensitizer.
Collapse
Affiliation(s)
- Joshua U-Jin Cheah
- NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, 21 Lower Kent Ridge, University Hall, Tan Chin Tuan Wing, #04-02, 119077, Singapore
| | - Heng Boon Low
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Blk MD4, Level 3, 117545, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, 28 Medical Drive, Centre for Life Sciences, Level 3, 117456, Singapore
| | - Yongliang Zhang
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Blk MD4, Level 3, 117545, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, 28 Medical Drive, Centre for Life Sciences, Level 3, 117456, Singapore
| | - James Chen Yong Kah
- NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, 21 Lower Kent Ridge, University Hall, Tan Chin Tuan Wing, #04-02, 119077, Singapore.,Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Blk E4, #04-08, 117583, Singapore
| |
Collapse
|
19
|
Novio F. Design of Targeted Nanostructured Coordination Polymers (NCPs) for Cancer Therapy. Molecules 2020; 25:E3449. [PMID: 32751178 PMCID: PMC7436016 DOI: 10.3390/molecules25153449] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
Conventional cancer chemotherapy presents notable drug side effects due to non-selective action of the chemotherapeutics to normal cells. Nanoparticles decorated with receptor-specific ligands on the surface have shown an important role in improving site-selective binding, retention, and drug delivery to the cancer cells. This review summarizes the recent reported achievements using nanostructured coordination polymers (NCPs) with active targeting properties for cancer treatment in vitro and in vivo. Despite the controversy surrounding the effectivity of active targeting nanoparticles, several studies suggest that active targeting nanoparticles notably increase the selectivity and the cytotoxic effect in tumoral cells over the conventional anticancer drugs and non-targeted nanoparticle platform, which enhances drug efficacy and safety. In most cases, the nanocarriers have been endowed with remarkable capabilities such as stimuli-responsive properties, targeting abilities, or the possibility to be monitored by imaging techniques. Unfortunately, the lack of preclinical studies impedes the evaluation of these unique and promising findings for the translation of NCPs into clinical trials.
Collapse
Affiliation(s)
- Fernando Novio
- Departament de Química, Universitat Autónoma de Barcelona, Campus UAB, Cerdanyola del Vallès, 08193 Barcelona, Spain
| |
Collapse
|
20
|
Combinatorial Inhibition of Cell Surface Receptors Using Dual Aptamer-Functionalized Nanoconstructs for Cancer Treatment. Pharmaceutics 2020; 12:pharmaceutics12070689. [PMID: 32708267 PMCID: PMC7408393 DOI: 10.3390/pharmaceutics12070689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/17/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Membrane receptors overexpressed in diseased states are considered novel therapeutic targets. However, the single targeting approach faces several fundamental issues, such as poor efficacy, resistance, and toxicity. Here, we report a dual-targeting strategy to enhance anti-cancer efficacy via synergistic proximity interactions between therapeutics and two receptor proteins. Importantly, we report the first finding of an interaction between c-Met and nucleolin and demonstrate the therapeutic value of targeting the interaction between them. Bispecific nanocarriers densely grafted with anti-c-Met and -nucleolin aptamer increased the local concentration of aptamers at the target sites, in addition to inducing target receptor clustering. It was also demonstrated that the simultaneous targeting of c-Met and nucleolin inhibited the cellular functions of the receptors and increased anti-cancer efficacy by altering the cell cycle. Our findings pave the way for the development of an effective combinatorial treatment based on nanoconstruct-mediated interaction between receptors.
Collapse
|
21
|
Mu J, Zhong H, Zou H, Liu T, Yu N, Zhang X, Xu Z, Chen Z, Guo S. Acid-sensitive PEGylated paclitaxel prodrug nanoparticles for cancer therapy: Effect of PEG length on antitumor efficacy. J Control Release 2020; 326:265-275. [PMID: 32687940 DOI: 10.1016/j.jconrel.2020.07.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/30/2020] [Accepted: 07/13/2020] [Indexed: 12/27/2022]
Abstract
Paclitaxel is one of the most widely used anticancer agents, but strong side effects and low bioavailability limit its clinical efficacy. The use of tumor microenvironment-responsive prodrugs is promising to solve these problems, and a smart linkage is crucial to achieve the efficient release of paclitaxel from such prodrugs in tumor. Herein, an acid-responsive acetone-based acyclic ketal linkage is used to construct paclitaxel prodrugs with different length of poly(ethylene glycol) (PEG). The PEGylated acetone-based acyclic-ketal-linked prodrugs of paclitaxel (PKPs) self-assembled into nanoparticles that were stable in normal physiological environment but released paclitaxel rapidly in mildly acidic environment in tumor. The length of PEG had considerable impact on size and critical micelle concentration of PKP nanoparticles, thereby affecting prodrug hydrolysis kinetics, pharmacokinetics, biodistribution, and antitumor activity for PKP nanoparticles. In an A2780 xenograft mouse model, PKP nanoparticles displayed improved pharmacokinetics and superior antitumor efficacy against Taxol. Our results demonstrate that acyclic-ketal-based prodrugs are useful for the development of acid-responsive anticancer nanomedicines.
Collapse
Affiliation(s)
- Jingqing Mu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Haiping Zhong
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Hui Zou
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Tao Liu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Na Yu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xi Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zunkai Xu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Ziqi Chen
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China..
| |
Collapse
|
22
|
Pearce KM, Okon I, Watson-Wright C. Induction of Oxidative DNA Damage and Epithelial Mesenchymal Transitions in Small Airway Epithelial Cells Exposed to Cosmetic Aerosols. Toxicol Sci 2020; 177:248-262. [DOI: 10.1093/toxsci/kfaa089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Abstract
Engineered metal nanoparticles (ENPs) are frequently incorporated into aerosolized consumer products, known as nano-enabled products (NEPs). Concern for consumer pulmonary exposures grows as NEPs produce high concentrations of chemically modified ENPs. A significant knowledge gap still exists surrounding NEP aerosol respiratory effects as previous research focuses on pristine/unmodified ENPs. Our research evaluated metal-containing aerosols emitted from nano-enabled cosmetics and their induction of oxidative stress and DNA damage, which may contribute to epithelial mesenchymal transitions (EMT) within primary human small airway epithelial cells. We utilized an automated NEP generation system to monitor and gravimetrically collect aerosols from two aerosolized cosmetic lines. Aerosol monitoring data were inputted into modeling software to determine potential inhaled dose and in vitro concentrations. Toxicological profiles of aerosols and comparable pristine ENPs (TiO2 and Fe2O3) were used to assess reactive oxygen species and oxidative stress by fluorescent-based assays. Single-stranded DNA (ssDNA) damage and 8-oxoguanine were detected using the CometChip assay after 24-h exposure. Western blots were conducted after 21-day exposure to evaluate modulation of EMT markers. Results indicated aerosols possessed primarily ultrafine particles largely depositing in tracheobronchial lung regions. Significant increases in oxidative stress, ssDNA damage, and 8-oxoguanine were detected post-exposure to aerosols versus pristine ENPs. Western blots revealed statistically significant decreases in E-cadherin and increases in vimentin, fascin, and CD44 for two aerosols, indicating EMT. This work suggests certain prolonged NEP inhalation exposures cause oxidative DNA damage, which may play a role in cellular changes associated with reduced respiratory function and should be of concern.
Collapse
Affiliation(s)
| | - Imoh Okon
- Center for Molecular & Translational Medicine, Georgia State University, Atlanta, Georgia 30302
| | | |
Collapse
|
23
|
PEGylated-Paclitaxel and Dihydroartemisinin Nanoparticles for Simultaneously Delivering Paclitaxel and Dihydroartemisinin to Colorectal Cancer. Pharm Res 2020; 37:129. [DOI: 10.1007/s11095-020-02819-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/07/2020] [Indexed: 12/17/2022]
|
24
|
Price E, Gesquiere AJ. Animal simulations facilitate smart drug design through prediction of nanomaterial transport to individual tissue cells. SCIENCE ADVANCES 2020; 6:eaax2642. [PMID: 32076633 PMCID: PMC7002136 DOI: 10.1126/sciadv.aax2642] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 11/25/2019] [Indexed: 05/27/2023]
Abstract
Smart drug design for antibody and nanomaterial-based therapies allows optimization of drug efficacy and more efficient early-stage preclinical trials. The ideal drug must display maximum efficacy at target tissue sites, with transport from tissue vasculature to the cellular environment being critical. Biological simulations, when coupled with in vitro approaches, can predict this exposure in a rapid and efficient manner. As a result, it becomes possible to predict drug biodistribution within single cells of live animal tissue without the need for animal studies. Here, we successfully utilized an in vitro assay and a computational fluid dynamic model to translate in vitro cell kinetics (accounting for cell-induced degradation) to whole-body simulations for multiple species as well as nanomaterial types to predict drug distribution into individual tissue cells. We expect this work to assist in refining, reducing, and replacing animal testing, while providing scientists with a new perspective during the drug development process.
Collapse
Affiliation(s)
- Edward Price
- Department of Chemistry, University of Central Florida, Orlando, FL 32816, USA
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Andre J. Gesquiere
- Department of Chemistry, University of Central Florida, Orlando, FL 32816, USA
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
- The College of Optics and Photonics (CREOL), University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
25
|
Sieber S, Grossen P, Bussmann J, Campbell F, Kros A, Witzigmann D, Huwyler J. Zebrafish as a preclinical in vivo screening model for nanomedicines. Adv Drug Deliv Rev 2019; 151-152:152-168. [PMID: 30615917 DOI: 10.1016/j.addr.2019.01.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/23/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022]
Abstract
The interactions of nanomedicines with biological environments is heavily influenced by their physicochemical properties. Formulation design and optimization are therefore key steps towards successful nanomedicine development. Unfortunately, detailed assessment of nanomedicine formulations, at a macromolecular level, in rodents is severely limited by the restricted imaging possibilities within these animals. Moreover, rodent in vivo studies are time consuming and expensive, limiting the number of formulations that can be practically assessed in any one study. Consequently, screening and optimisation of nanomedicine formulations is most commonly performed in surrogate biological model systems, such as human-derived cell cultures. However, despite the time and cost advantages of classical in vitro models, these artificial systems fail to reflect and mimic the complex biological situation a nanomedicine will encounter in vivo. This has acutely hampered the selection of potentially successful nanomedicines for subsequent rodent in vivo studies. Recently, zebrafish have emerged as a promising in vivo model, within nanomedicine development pipelines, by offering opportunities to quickly screen nanomedicines under in vivo conditions and in a cost-effective manner so as to bridge the current gap between in vitro and rodent studies. In this review, we outline several advantageous features of the zebrafish model, such as biological conservation, imaging modalities, availability of genetic tools and disease models, as well as their various applications in nanomedicine development. Critical experimental parameters are discussed and the most beneficial applications of the zebrafish model, in the context of nanomedicine development, are highlighted.
Collapse
Affiliation(s)
- Sandro Sieber
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Jeroen Bussmann
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Frederick Campbell
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Alexander Kros
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Department of Biochemistry and Molecular Biology, University of British Columbia, Health Sciences Mall, Vancouver, British Columbia, Canada..
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
26
|
Ngo N, Choucair K, Creeden JF, Qaqish H, Bhavsar K, Murphy C, Lian K, Albrethsen MT, Stanbery L, Phinney RC, Brunicardi FC, Dworkin L, Nemunaitis J. Bifidobacterium spp: the promising Trojan Horse in the era of precision oncology. Future Oncol 2019; 15:3861-3876. [PMID: 31668087 DOI: 10.2217/fon-2019-0374] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Selective delivery of therapeutic agents into solid tumors has been a major challenge impeding the achievement of long-term disease remission and cure. The need to develop alternative drug delivery routes to achieve higher drug concentration in tumor tissue, reduce unwanted off-target side effects and thus achieve greater therapeutic efficacy, has resulted in an explosive body of research. Bifidobacterium spp. are anaerobic, nonpathogenic, Gram-positive bacteria, commensal to the human gut that are a possible anticancer drug-delivery vehicle. In this review, we describe Bifidobacterium's microbiology, current clinical applications, overview of the preclinical work investigating Bifidobacterium's potential to deliver anticancer therapy, and review the different strategies used up to date. Finally, we discuss both current challenges and future prospects.
Collapse
Affiliation(s)
- Nealie Ngo
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Khalil Choucair
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Justin F Creeden
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Hanan Qaqish
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Krupa Bhavsar
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Chantal Murphy
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Kendra Lian
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Mary T Albrethsen
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Laura Stanbery
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | | | - F Charles Brunicardi
- Department of Surgery, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Lance Dworkin
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - John Nemunaitis
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
- ProMedica Health System, Toledo, OH 43606, USA
| |
Collapse
|
27
|
Price E, Gesquiere AJ. An in vitro assay and artificial intelligence approach to determine rate constants of nanomaterial-cell interactions. Sci Rep 2019; 9:13943. [PMID: 31558741 PMCID: PMC6763461 DOI: 10.1038/s41598-019-50208-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022] Open
Abstract
In vitro assays and simulation technologies are powerful methodologies that can inform scientists of nanomaterial (NM) distribution and fate in humans or pre-clinical species. For small molecules, less animal data is often needed because there are a multitude of in vitro screening tools and simulation-based approaches to quantify uptake and deliver data that makes extrapolation to in vivo studies feasible. Small molecule simulations work because these materials often diffuse quickly and partition after reaching equilibrium shortly after dosing, but this cannot be applied to NMs. NMs interact with cells through energy dependent pathways, often taking hours or days to become fully internalized within the cellular environment. In vitro screening tools must capture these phenomena so that cell simulations built on mechanism-based models can deliver relationships between exposure dose and mechanistic biology, that is biology representative of fundamental processes involved in NM transport by cells (e.g. membrane adsorption and subsequent internalization). Here, we developed, validated, and applied the FORECAST method, a combination of a calibrated fluorescence assay (CF) with an artificial intelligence-based cell simulation to quantify rates descriptive of the time-dependent mechanistic biological interactions between NMs and individual cells. This work is expected to provide a means of extrapolation to pre-clinical or human biodistribution with cellular level resolution for NMs starting only from in vitro data.
Collapse
Affiliation(s)
- Edward Price
- NanoScience Technology Center, University of Central Florida, Orlando, FL, 32826, USA
- Department of Chemistry, University of Central Florida, Orlando, FL, 32816, USA
| | - Andre J Gesquiere
- NanoScience Technology Center, University of Central Florida, Orlando, FL, 32826, USA.
- Department of Chemistry, University of Central Florida, Orlando, FL, 32816, USA.
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, 32816, USA.
- The College of Optics and Photonics (CREOL), University of Central Florida, Orlando, FL, 32816, USA.
| |
Collapse
|
28
|
Tulbah AS, Pisano E, Landh E, Scalia S, Young PM, Traini D, Ong HX. Simvastatin Nanoparticles Reduce Inflammation in LPS-Stimulated Alveolar Macrophages. J Pharm Sci 2019; 108:3890-3897. [PMID: 31494116 DOI: 10.1016/j.xphs.2019.08.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023]
Abstract
Simvastatin (SV) is widely used as a lipid-lowering medication that has also been found to have beneficial immunomodulatory effects for treatment of chronic lung diseases. Although its anti-inflammatory activity has been investigated, its underlying mechanisms have not yet been clearly elucidated. In this study, the anti-inflammatory and antioxidant effects and mechanism of simvastatin nanoparticles (SV-NPs) on lipopolysaccharide-stimulated alveolar macrophages (AMs) NR8383 cells were investigated. Quantitative cellular uptake of SV-NPs, the production of inflammatory mediators (interleukin-6, tumor necrosis factor, and monocyte chemoattractant protein-1), and oxidative stress (nitric oxide) were tested. Furthermore, the involvement of the nuclear factor κB (NF-κB) signaling pathway in activation of inflammation in AMs and the efficacy of SV were visualized using immunofluorescence. Results indicated that SV-NPs exhibit a potent inhibitory effect on nitric oxide production and secretion of inflammatory cytokine in inflamed AM, without affecting cell viability. The enhanced anti-inflammatory activity of SV-NPs is likely due to SV-improved chemical-physical stability and higher cellular uptake into AM. The study also indicates that SV targets the inflammatory and oxidative response of AM, through inactivation of the NF-κB signaling pathway, supporting the pharmacological basis of SV for treatment of chronic inflammatory lung diseases.
Collapse
Affiliation(s)
- Alaa S Tulbah
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, 2037 New South Wales, Australia; College of Pharmacy, Umm Al Qura University, Makkah, Saudi Arabia
| | - Elvira Pisano
- Dipartimento di Scienze della vita e biotecnologie, University of Ferrara, Italy
| | - Emelie Landh
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, 2037 New South Wales, Australia
| | - Santo Scalia
- Dipartimento di Scienze della vita e biotecnologie, University of Ferrara, Italy
| | - Paul M Young
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, 2037 New South Wales, Australia
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, 2037 New South Wales, Australia
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, 2037 New South Wales, Australia.
| |
Collapse
|
29
|
Shahjin F, Chand S, Yelamanchili SV. Extracellular Vesicles as Drug Delivery Vehicles to the Central Nervous System. J Neuroimmune Pharmacol 2019; 15:443-458. [DOI: 10.1007/s11481-019-09875-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023]
|
30
|
da Rocha FR, Haupenthal DPDS, Zaccaron RP, Corrêa MEAB, Tramontin NDS, Fonseca JP, Nesi RT, Muller AP, Pinho RA, Paula MMDS, Silveira PCL. Therapeutic effects of iontophoresis with gold nanoparticles in the repair of traumatic muscle injury. J Drug Target 2019; 28:307-319. [PMID: 31379221 DOI: 10.1080/1061186x.2019.1652617] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Studies have shown the benefits of gold nanoparticles (GNPs) in muscle and epithelial injury models. In physiotherapy, the use of the microcurrent apparatus is associated with certain drugs (Iontophoresis) to increase the topical penetration and to associate the effects of both therapies. Therefore, the objective of this study was to investigate the effects of iontophoresis along with GNPs in the skeletal muscle of rats exposed to a traumatic muscle injury. We utilised 50 Wistar rats randomly divided in to five experimental groups (n = 10): Control group (CG); Muscle injury group (MI); MI + GNPs (20 nm, 30 mg kg-1); MI + Microcurrent (300 μA); and MI + Microcurrent + GNPs. The treatment was performed daily for 7 days, with the first session starting at 24 h after the muscle injury. The animals were sacrificed and the gastrocnemius muscle was surgically removedand stored for the proper evaluations. The group that received iontophoresis with GNPs showed significant differences in inflammation and oxidative stress parameters and in the histopathological evaluation showed preserved morphology. In addition, we observed an improvement in the locomotor response and pain symptoms of these animals. These results suggest that the association of boththerapies accelerates the inflammatory response of the injured limb.
Collapse
Affiliation(s)
- Franciani Rodrigues da Rocha
- Laboratory of Experimental Phisiopatology, Program of Post Graduate in Science of Health, Universidade Do Extremo Sul Catarinense, Criciúma, Brazil
| | - Daniela Pacheco Dos Santo Haupenthal
- Laboratory of Experimental Phisiopatology, Program of Post Graduate in Science of Health, Universidade Do Extremo Sul Catarinense, Criciúma, Brazil
| | - Rubya Pereira Zaccaron
- Laboratory of Experimental Phisiopatology, Program of Post Graduate in Science of Health, Universidade Do Extremo Sul Catarinense, Criciúma, Brazil
| | | | - Natalia Dos Santos Tramontin
- Laboratory of Translacional Biomedicine, Program of Post Graduate in Science of Health, Universidade Do Extremo Sul Catarinense, Criciúma, Brazil
| | - Jeandro Paes Fonseca
- Postgraduate Program in Materials Science and Engineering, Federal Amazonas University, Manaus-Amazonas, Brazil
| | - Renata Tiscoski Nesi
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica Do Paraná, Curitiba, Brazil
| | - Alexandre Pastoris Muller
- Laboratory of Translacional Biomedicine, Program of Post Graduate in Science of Health, Universidade Do Extremo Sul Catarinense, Criciúma, Brazil
| | - Ricardo Aurino Pinho
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica Do Paraná, Curitiba, Brazil
| | | | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Phisiopatology, Program of Post Graduate in Science of Health, Universidade Do Extremo Sul Catarinense, Criciúma, Brazil
| |
Collapse
|
31
|
I El-Gogary R, Gaber SAA, Nasr M. Polymeric nanocapsular baicalin: Chemometric optimization, physicochemical characterization and mechanistic anticancer approaches on breast cancer cell lines. Sci Rep 2019; 9:11064. [PMID: 31363132 PMCID: PMC6667692 DOI: 10.1038/s41598-019-47586-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 07/19/2019] [Indexed: 01/12/2023] Open
Abstract
Baicalin is a multi-purpose flavonoid known for its anticancer properties, but its application is hindered by its low water solubility and bioavailability. Polymeric nanocapsules were proposed in this work as a promising system for enhancing baicalin delivery, and potentiating its anticancer properties. The characterization of nanocapsules was augmented with chemometric analysis, and the selected formulations were tested on two breast cancer cell lines (MCF-7 and MDA-MB-231), with mechanistic anticancer elucidation using MTT assay, confocal microscopy uptake, flow cytometry, mechanism of cell death, reactive oxygen species production, caspase 3/7 activity and death biomarker expression using quantitative real time PCR. Results showed that baicalin nanocapsules displayed favorable pharmaceutical properties; with the formulation variables affecting their properties elucidated using chemometric factorial analysis. Nanocapsules enhanced the anticancer activity of baicalin up to 216 times for MCF-7 cells and 31 times for MDA-MB-231 after 24 hr incubation. Cellular internalization of the fluorescently labeled nanocapsules was confirmed after 4 hr incubation for both cell lines. Apoptosis was the dominant cell death mechanism, with significant up-regulation of P53 in baicalin nanocapsules treated cells. Data here presented drive to further preclinical studies to investigate the delivery of baicalin polymeric nanocapsules and their anti-cancer activity.
Collapse
Affiliation(s)
- Riham I El-Gogary
- Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sara A Abdel Gaber
- Nanomedicine Department, Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
32
|
Misra SK, Wu Z, Ostadhossein F, Ye M, Boateng K, Schulten K, Tajkhorshid E, Pan D. Pro-Nifuroxazide Self-Assembly Leads to Triggerable Nanomedicine for Anti-cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:18074-18089. [PMID: 31013055 PMCID: PMC7066988 DOI: 10.1021/acsami.9b01343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Transcription factor STAT3 has been shown to regulate genes that are involved in stem cell self-renewal and thus represents a novel therapeutic target of great biological significance. However, many small-molecule agents with potential effects through STAT3 modulation in cancer therapy lack aqueous solubility and high off-target toxicity, hence impeding efficient bioavailability and activity. This work, for the first time, reports a prodrug-based strategy for selective and safer delivery of STAT3 inhibitors designed toward metastatic and drug-resistant breast cancer. We have synthesized a novel lipase-labile SN-2 phospholipid prodrug from a clinically investigated STAT3 inhibitor, nifuroxazide (Pro-nifuroxazide), which can be regioselectively cleaved by the membrane-abundant enzymes in cancer cells. Pro-nifuroxazide self-assembled to sub 20 nm nanoparticles (NPs), and the cytotoxic ability was screened in ER(+)-MCF-7 and ER(-)-MD-MB231 cells at 48-72 h using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetra-zolium bromide proliferation assay. Results indicated that Pro-nifuroxazide NPs are multifold more effective toward inhibiting cancer cells in a time-dependent manner compared to parent nifuroxazide. A remarkable improvement in the local concentration of drugs to as high as ∼240 fold when assembled into NPs is presumably the reason for this functional improvement. We also introduced molecular dynamics simulations to generate Pro-nifuroxazide nano-assembly, as a model assembly from triggerable anti-cancer drugs, to provide molecular insights correlating physicochemical and anti-cancer properties. In silico properties of Pro-nifuroxazide including size, chemistry of NPs and membrane interactions with individual molecules could be validated by in vitro functional activities in cells of breast cancer origin. The in vivo anti-cancer efficiencies of Pro-nifuroxazide NPs in nude mice xenografts with MCF-7 revealed remarkable growth inhibition of as high as 400%. Histopathological analysis corroborated these findings to show significantly high nuclear fragmentation and retracted cytoplasm. Immunostaining on tumor section demonstrated a significantly lower level of pSTAT-3 by Pro-nifuroxazide NP treatment, establishing the inhibition of STAT-3 phosphorylation. Our strategy for the first time proposes a translatable prodrug agent self-assembled into NPs and demonstrates remarkable enhancement in IC50, induced apoptosis, and reduced cancer cell population through STAT-3 inhibition via reduced phosphorylation.
Collapse
Affiliation(s)
- Santosh K Misra
- Department of Bioengineering , University of Illinois at Urbana-Champaign , Urbana 61801 , United States
- Mills Breast Cancer Institute, Carle Foundation Hospital , 502 N. Busey , Urbana , Illinois 61801 , United States
| | | | - Fatemeh Ostadhossein
- Department of Bioengineering , University of Illinois at Urbana-Champaign , Urbana 61801 , United States
- Mills Breast Cancer Institute, Carle Foundation Hospital , 502 N. Busey , Urbana , Illinois 61801 , United States
| | - Mao Ye
- Department of Bioengineering , University of Illinois at Urbana-Champaign , Urbana 61801 , United States
- Mills Breast Cancer Institute, Carle Foundation Hospital , 502 N. Busey , Urbana , Illinois 61801 , United States
| | | | | | | | - Dipanjan Pan
- Department of Bioengineering , University of Illinois at Urbana-Champaign , Urbana 61801 , United States
- Mills Breast Cancer Institute, Carle Foundation Hospital , 502 N. Busey , Urbana , Illinois 61801 , United States
| |
Collapse
|
33
|
Choi DH, Park YS. Arginine-rich Peptide Coated PLGA Nanoparticles Enhance Polymeric Delivery of Antisense HIF1α-oligonucleotide to Fully Differentiated Stiff Adipocytes. TOXICOLOGY AND ENVIRONMENTAL HEALTH SCIENCES 2019; 11:1-10. [DOI: 10.1007/s13530-019-0382-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/16/2018] [Accepted: 12/26/2018] [Indexed: 01/05/2025]
|
34
|
Pereira SGT, Hudoklin S, Kreft ME, Kostevsek N, Stuart MCA, Al-Jamal WT. Intracellular Activation of a Prostate Specific Antigen-Cleavable Doxorubicin Prodrug: A Key Feature Toward Prodrug-Nanomedicine Design. Mol Pharm 2019; 16:1573-1585. [DOI: 10.1021/acs.molpharmaceut.8b01257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sara G. T. Pereira
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, U.K
| | - Samo Hudoklin
- University of Ljubljana, Faculty of Medicine, Institute of Cell Biology, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- University of Ljubljana, Faculty of Medicine, Institute of Cell Biology, Ljubljana, Slovenia
| | - Nina Kostevsek
- Department for Nanostructured Materials, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Marc C. A. Stuart
- Electron Microscopy, University of Groningen, Nijenborgh 7, 9747AG Groningen, The Netherlands
| | - Wafa T. Al-Jamal
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, U.K
| |
Collapse
|
35
|
Sorasitthiyanukarn FN, Muangnoi C, Ratnatilaka Na Bhuket P, Rojsitthisak P, Rojsitthisak P. Chitosan/alginate nanoparticles as a promising approach for oral delivery of curcumin diglutaric acid for cancer treatment. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 93:178-190. [DOI: 10.1016/j.msec.2018.07.069] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 06/25/2018] [Accepted: 07/24/2018] [Indexed: 12/21/2022]
|
36
|
Argudo PG, Carril M, Martín-Romero MT, Giner-Casares JJ, Carrillo-Carrión C. Surface-Active Fluorinated Quantum Dots for Enhanced Cellular Uptake. Chemistry 2018; 25:195-199. [PMID: 30257052 DOI: 10.1002/chem.201804704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Indexed: 12/28/2022]
Abstract
Fluorescent nanoparticles, such as quantum dots, hold great potential for biomedical applications, mainly sensing and bioimaging. However, the inefficient cell uptake of some nanoparticles hampers their application in clinical practice. Here, the effect of the modification of the quantum dot surface with fluorinated ligands to increase their surface activity and, thus, enhance their cellular uptake was explored.
Collapse
Affiliation(s)
- Pablo G Argudo
- Institute of Fine Chemistry and Nanochemistry, Department of Physical Chemistry and Applied Thermodynamics, University of Córdoba, Campus de Rabanales, Edificio Marie Curie, 14014, Córdoba, Spain
| | - Mónica Carril
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country UPV/EHU, Barrio Sarriena s/n, 48940, Leioa, Spain.,Ikerbasque, Basque Foundation for Science, Maria Diaz de Haro 3,6 solaiura, 48011, Bilbao, Spain
| | - María T Martín-Romero
- Institute of Fine Chemistry and Nanochemistry, Department of Physical Chemistry and Applied Thermodynamics, University of Córdoba, Campus de Rabanales, Edificio Marie Curie, 14014, Córdoba, Spain
| | - Juan J Giner-Casares
- Institute of Fine Chemistry and Nanochemistry, Department of Physical Chemistry and Applied Thermodynamics, University of Córdoba, Campus de Rabanales, Edificio Marie Curie, 14014, Córdoba, Spain
| | - Carolina Carrillo-Carrión
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Paseo Miramón 182, 20014, San Sebastian, Spain.,Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, Jenaro de la Fuente s/n, 15782, Santiago de Compostela, Spain
| |
Collapse
|
37
|
Gupta B, Poudel BK, Ruttala HB, Regmi S, Pathak S, Gautam M, Jin SG, Jeong JH, Choi HG, Ku SK, Yong CS, Kim JO. Hyaluronic acid-capped compact silica-supported mesoporous titania nanoparticles for ligand-directed delivery of doxorubicin. Acta Biomater 2018; 80:364-377. [PMID: 30201431 DOI: 10.1016/j.actbio.2018.09.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/04/2018] [Accepted: 09/06/2018] [Indexed: 12/26/2022]
Abstract
Mesoporous titania nanoparticles (MTN), owing to their high surface area to volume ratio and tunable pore sizes, appear capable of delivering sizable amounts of drug payloads, and hence, show considerable promise as drug delivery candidates in cancer therapy. We designed silica-supported MTN (MTNst) coated with hyaluronic acid (HA) to effectively deliver doxorubicin (DOX) for breast cancer therapy. The HA coating served a dual purpose of stabilizing the payload in the carriers as well as actively targeting the nanodevices to CD44 receptors. The so-formed HA-coated MTNst carrying DOX (HA/DOX-MTNst) had spheroid particles with a considerable drug-loading capacity and showed significantly superior in vitro cytotoxicity against MDA-MB-231 cells as compared to free DOX. HA/DOX-MTNst markedly improved the cellular uptake of DOX in an apparently CD44 receptor-dependent manner, and increased the number of apoptotic cells as compared to free DOX. These nanoplatforms accumulated in large quantities in the tumors of MDA-MB-231 xenograft tumor-bearing mice, where they significantly enhanced the inhibition of tumor growth compared to that observed with free DOX with no signs of acute toxicity. Based on these excellent results, we deduced that HA/DOX-MTNst could be successfully used for targeted breast cancer therapy. STATEMENT OF SIGNIFICANCE: This is the first study to use silica-supported mesoporous titania nanoparticles (MTNst) for doxorubicin (DOX) delivery to treat breast cancer, which exhibited effective and enhanced in vitro and in vivo apoptosis and tumor growth inhibition. Solid silica was used to support the mesoporous TiO2 resulting in MTNst, which efficiently incorporated a high DOX payload. The hyaluronic acid (HA) coating over the MTNst surface served a dual purpose of first, stabilizing DOX inside the MTNst (capping agent), and second, directing the nanoplatform device to CD44 receptors that are highly expressed in MDA-MB-231 cells (targeting ligand). The NPs exhibited highly efficacious in vitro tumor-cell killing and excellent in vivo tumor regression, highlighting the enormous promise of this system for breast cancer therapy.
Collapse
Affiliation(s)
- Biki Gupta
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, Republic of Korea
| | - Bijay Kumar Poudel
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, Republic of Korea
| | - Hima Bindu Ruttala
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, Republic of Korea
| | - Shobha Regmi
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, Republic of Korea
| | - Shiva Pathak
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, Republic of Korea
| | - Milan Gautam
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, Republic of Korea
| | - Sung Giu Jin
- Department of Pharmaceutical Engineering, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan 31116, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan 712-702, Republic of Korea.
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, Republic of Korea.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, Republic of Korea.
| |
Collapse
|
38
|
Kauffman AC, Piotrowski-Daspit AS, Nakazawa KH, Jiang Y, Datye A, Saltzman WM. Tunability of Biodegradable Poly(amine- co-ester) Polymers for Customized Nucleic Acid Delivery and Other Biomedical Applications. Biomacromolecules 2018; 19:3861-3873. [PMID: 30110158 PMCID: PMC6510397 DOI: 10.1021/acs.biomac.8b00997] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Gene therapy promises to treat diseases that arise from genetic abnormalities by correcting the underlying cause of the disease rather than treating the associated symptoms. Successful transfer of nucleic acids into cells requires efficient delivery vehicles that protect the cargo and can penetrate the appropriate cellular barriers before releasing their contents. Many viral vectors and synthetic polycationic vectors for nucleic acid delivery do not translate well from in vitro to in vivo applications due to their instability and toxicity. We synthesized and characterized a library of biocompatible low charge density polymers from a family of poly(amine- co-ester) (PACE) terpolymers produced via enzyme catalyzed polymerization. PACE polymers are highly customizable; we found that the terpolymer composition can be optimized to produce efficient transfection of various nucleic acids-including DNA plasmids, mRNA, and siRNA-in specific cell types with low toxicity. Our findings suggest that the unique tunability of PACEs offers new tools for gene therapy and other biomedical applications.
Collapse
Affiliation(s)
- Amy C. Kauffman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| | | | - Kay H. Nakazawa
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| | - Yuhang Jiang
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| | - Amit Datye
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06511
| | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT 06511
- Department of Physiology, Yale University, New Haven, CT 06511
| |
Collapse
|
39
|
Huth K, Glaeske M, Achazi K, Gordeev G, Kumar S, Arenal R, Sharma SK, Adeli M, Setaro A, Reich S, Haag R. Fluorescent Polymer-Single-Walled Carbon Nanotube Complexes with Charged and Noncharged Dendronized Perylene Bisimides for Bioimaging Studies. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1800796. [PMID: 29870583 DOI: 10.1002/smll.201800796] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/20/2018] [Indexed: 05/28/2023]
Abstract
Fluorescent nanomaterials are expected to revolutionize medical diagnostic, imaging, and therapeutic tools due to their superior optical and structural properties. Their inefficient water solubility, cell permeability, biodistribution, and high toxicity, however, limit the full potential of their application. To overcome these obstacles, a water-soluble, fluorescent, cytocompatible polymer-single-walled carbon nanotube (SWNT) complex is introduced for bioimaging applications. The supramolecular complex consists of an alkylated polymer conjugated with neutral hydroxylated or charged sulfated dendronized perylene bisimides (PBIs) and SWNTs as a general immobilization platform. The polymer backbone solubilizes the SWNTs, decorates them with fluorescent PBIs, and strongly improves their cytocompatibility by wrapping around the SWNT scaffold. In photophysical measurements and biological in vitro studies, sulfated complexes exhibit superior optical properties, cellular uptake, and intracellular staining over their hydroxylated analogs. A toxicity assay confirms the highly improved cytocompatibility of the polymer-wrapped SWNTs toward surfactant-solubilized SWNTs. In microscopy studies the complexes allow for the direct imaging of the SWNTs' cellular uptake via the PBI and SWNT emission using the 1st and 2nd optical window for bioimaging. These findings render the polymer-SWNT complexes with nanometer size, dual fluorescence, multiple charges, and high cytocompatibility as valuable systems for a broad range of fluorescence bioimaging studies.
Collapse
Affiliation(s)
- Katharina Huth
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Mareen Glaeske
- Department of Physics, Freie Universität Berlin, 14195, Berlin, Germany
| | - Katharina Achazi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Georgy Gordeev
- Department of Physics, Freie Universität Berlin, 14195, Berlin, Germany
| | - Shiv Kumar
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Raúl Arenal
- Institute of Nanoscience of Aragon (INA), Advanced Microscopy Laboratory (LMA), University of Zaragoza, 50018, Zaragoza, Spain
- Foundation ARAID, 50018, Zaragoza, Spain
| | - Sunil K Sharma
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Mohsen Adeli
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
- Department of Chemistry, Faculty of Science, Lorestan University, Khorram Abad, 68151-44316, Iran
| | - Antonio Setaro
- Department of Physics, Freie Universität Berlin, 14195, Berlin, Germany
| | - Stephanie Reich
- Department of Physics, Freie Universität Berlin, 14195, Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| |
Collapse
|
40
|
Jardim KV, Palomec-Garfias AF, Andrade BYG, Chaker JA, Báo SN, Márquez-Beltrán C, Moya SE, Parize AL, Sousa MH. Novel magneto-responsive nanoplatforms based on MnFe 2O 4 nanoparticles layer-by-layer functionalized with chitosan and sodium alginate for magnetic controlled release of curcumin. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:184-195. [PMID: 30184741 DOI: 10.1016/j.msec.2018.06.039] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 05/22/2018] [Accepted: 06/18/2018] [Indexed: 02/08/2023]
Abstract
Remotely assisted drug delivery by means of magnetic biopolymeric nanoplatforms has been utilized as an important tool to improve the delivery/release of hydrophobic drugs and to address their low cargo capacity. In this work, MnFe2O4 magnetic nanoparticles (MNPs) were synthesized by thermal decomposition, coated with citrate and then functionalized with the layer-by-layer (LbL) assembly of polyelectrolyte multilayers, with chitosan as polycation and sodium alginate as polyanion. Simultaneous conductimetric and potentiometric titrations were employed to optimize the LbL deposition and to enhance the loading capacity of nanoplatforms for curcumin, a hydrophobic drug used in cancer treatment. ~200 nm sized biopolymer platforms with ~12 nm homogeneously embedded MNPs were obtained and characterized by means of XRD, HRTEM, DLS, TGA, FTIR, XPS and fluorescence spectroscopy techniques to access structural, morphological and surface properties, to probe biopolymer functionalization and to quantify drug-loading. Charge reversals (±30 mV) after each deposition confirmed polyelectrolyte adsorption and a stable LbL assembly. Magnetic interparticle interaction was reduced in the biopolymeric structure, hinting at an optimized performance in magnetic hyperthermia for magneto-assisted drug release applications. Curcumin was encapsulated, resulting in an enhanced payload (~100 μg/mg). Nanocytotoxicity assays showed that the biopolymer capping enhanced the biocompatibility of nanoplatforms, maintaining entrapped curcumin. Our results indicate the potential of synthesized nanoplatforms as an alternative way of remotely delivering/releasing curcumin for medical purposes, upon application of an alternating magnetic field, demonstrating improved efficiency and reduced toxicity.
Collapse
Affiliation(s)
| | | | - Bárbara Yasmin Garcia Andrade
- Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil
| | | | - Sônia Nair Báo
- Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil
| | - César Márquez-Beltrán
- Instituto de Física - Benemérita Universidad Autónoma de Puebla, Puebla, Pue 72570, Mexico
| | - Sergio Enrique Moya
- Soft Matter Nanotechnology Laboratory, CIC biomaGUNE, San Sebastián, Guip 20009, Spain
| | - Alexandre Luis Parize
- Polimat, Grupo de Estudos em Materiais Poliméricos, Departamento de Química, Universidade Federal de Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | | |
Collapse
|
41
|
Angeloni L, Reggente M, Passeri D, Natali M, Rossi M. Identification of nanoparticles and nanosystems in biological matrices with scanning probe microscopy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 10:e1521. [PMID: 29665287 DOI: 10.1002/wnan.1521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 02/26/2018] [Accepted: 03/10/2018] [Indexed: 01/22/2023]
Abstract
Identification of nanoparticles and nanosystems into cells and biological matrices is a hot research topic in nanobiotechnologies. Because of their capability to map physical properties (mechanical, electric, magnetic, chemical, or optical), several scanning probe microscopy based techniques have been proposed for the subsurface detection of nanomaterials in biological systems. In particular, atomic force microscopy (AFM) can be used to reveal stiff nanoparticles in cells and other soft biomaterials by probing the sample mechanical properties through the acquisition of local indentation curves or through the combination of ultrasound-based methods, like contact resonance AFM (CR-AFM) or scanning near field ultrasound holography. Magnetic force microscopy can detect magnetic nanoparticles and other magnetic (bio)materials in nonmagnetic biological samples, while electric force microscopy, conductive AFM, and Kelvin probe force microscopy can reveal buried nanomaterials on the basis of the differences between their electric properties and those of the surrounding matrices. Finally, scanning near field optical microscopy and tip-enhanced Raman spectroscopy can visualize buried nanostructures on the basis of their optical and chemical properties. Despite at a still early stage, these methods are promising for detection of nanomaterials in biological systems as they could be truly noninvasive, would not require destructive and time-consuming specific sample preparation, could be performed in vitro, on alive samples and in water or physiological environment, and by continuously imaging the same sample could be used to dynamically monitor the diffusion paths and interaction mechanisms of nanomaterials into cells and biological systems. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Livia Angeloni
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Rome, Italy
| | - Melania Reggente
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Rome, Italy
| | - Daniele Passeri
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Rome, Italy
| | - Marco Natali
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Rome, Italy
| | - Marco Rossi
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Rome, Italy.,Research Center for Nanotechnology Applied to Engineering of Sapienza University of Rome (CNIS), Rome, Italy
| |
Collapse
|
42
|
Manatunga DC, de Silva RM, de Silva KMN, Malavige GN, Wijeratne DT, Williams GR, Jayasinghe CD, Udagama PV. Effective delivery of hydrophobic drugs to breast and liver cancer cells using a hybrid inorganic nanocarrier: A detailed investigation using cytotoxicity assays, fluorescence imaging and flow cytometry. Eur J Pharm Biopharm 2018; 128:18-26. [PMID: 29625162 DOI: 10.1016/j.ejpb.2018.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/30/2018] [Accepted: 04/02/2018] [Indexed: 02/07/2023]
Abstract
This study was focused on developing a drug carrier system composed of a polymer containing hydroxyapatite (HAp) shell and a magnetic core of iron oxide nanoparticles. Doxorubicin and/or curcumin were loaded into the carrier via a simple diffusion deposition approach, with encapsulation efficiencies (EE) for curcumin and doxorubicin of 93.03 ± 0.3% and 97.37 ± 0.12% respectively. The co-loading of curcumin and doxorubicin led to a total EE of 76.02 ± 0.48%. Release studies were carried out at pH 7.4 and 5.3, and revealed a greater extent of release at pH 5.3, showing the formulations to have potential applications in tumor microenvironments. Cytotoxicity assays, fluorescence imaging and flow cytometry demonstrated that the formulations could effectively inhibit the growth of MCF-7 (breast) and HEpG2 (liver) cancer cells, being more potent than the free drug molecules both in terms of dose and duration of action. Additionally, hemolysis tests and cytotoxicity evaluations determined the drug-loaded carriers to be non-toxic towards non-cancerous cells. These formulations thus have great potential in the development of new cancer therapeutics.
Collapse
Affiliation(s)
| | - Rohini M de Silva
- Department of Chemistry, University of Colombo, Colombo 00300, Sri Lanka.
| | - K M Nalin de Silva
- Department of Chemistry, University of Colombo, Colombo 00300, Sri Lanka; Sri Lanka Institute of Nanotechnology (SLINTEC), Nanotechnology & Science Park, Mahenwatte, Pitipana, Homagama 10206, Sri Lanka
| | - Gathsaurie Neelika Malavige
- Center for Dengue Research, Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayewardenepura, 10250, Sri Lanka
| | - Dulharie T Wijeratne
- Center for Dengue Research, Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayewardenepura, 10250, Sri Lanka
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | | | - Preethi V Udagama
- Department of Zoology, University of Colombo, Colombo 00300, Sri Lanka
| |
Collapse
|
43
|
Poynton FE, Bright SA, Blasco S, Williams DC, Kelly JM, Gunnlaugsson T. The development of ruthenium(ii) polypyridyl complexes and conjugates for in vitro cellular and in vivo applications. Chem Soc Rev 2018; 46:7706-7756. [PMID: 29177281 DOI: 10.1039/c7cs00680b] [Citation(s) in RCA: 315] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ruthenium(ii) [Ru(ii)] polypyridyl complexes have been the focus of intense investigations since work began exploring their supramolecular interactions with DNA. In recent years, there have been considerable efforts to translate this solution-based research into a biological environment with the intention of developing new classes of probes, luminescent imaging agents, therapeutics and theranostics. In only 10 years the field has expanded with diverse applications for these complexes as imaging agents and promising candidates for therapeutics. In light of these efforts this review exclusively focuses on the developments of these complexes in biological systems, both in cells and in vivo, and hopes to communicate to readers the diversity of applications within which these complexes have found use, as well as new insights gained along the way and challenges that researchers in this field still face.
Collapse
Affiliation(s)
- Fergus E Poynton
- School of Chemistry and Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, The University of Dublin, Dublin 2, Ireland.
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
The treatment of malignancies has undergone dramatic changes in the past few decades. Advances in drug delivery techniques and nanotechnology have allowed for new formulations of old drugs, so as to improve the pharmacokinetics, to enhance accumulation in solid tumors, and to reduce the significant toxic effects of these important therapeutic agents. Here, we review the published clinical data in cancer therapy of several major drug delivery systems, including targeted radionuclide therapy, antibody-drug conjugates, liposomes, polymer-drug conjugates, polymer implants, micelles, and nanoparticles. The clinical outcomes of these delivery systems from various phases of clinical trials are summarized. The success and limitations of the drug delivery strategies are discussed based on the clinical observations. In addition, the challenges in applying drug delivery for efficacious cancer therapy, including physical barriers, tumor heterogeneity, drug resistance, and metastasis, are discussed along with future perspectives of drug delivery in cancer therapy. In doing so, we intend to underscore that efficient delivery of cancer therapeutics to solid malignancies remains a major challenge in cancer therapy, and requires a multidisciplinary approach that integrates knowledge from the diverse fields of chemistry, biology, engineering, and medicine. The overall objective of this review is to improve our understanding of the clinical fate of commonly investigated drug delivery strategies, and to identify the limitations that must be addressed in future drug delivery strategies, toward the pursuit of curative therapies for cancer.
Collapse
Affiliation(s)
- Zheng-Rong Lu
- Case Center for Biomolecular Engineering, Department of Biomedical Engineering , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Peter Qiao
- Case Center for Biomolecular Engineering, Department of Biomedical Engineering , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| |
Collapse
|
45
|
Sieber S, Grossen P, Detampel P, Siegfried S, Witzigmann D, Huwyler J. Zebrafish as an early stage screening tool to study the systemic circulation of nanoparticulate drug delivery systems in vivo. J Control Release 2017; 264:180-191. [PMID: 28851572 DOI: 10.1016/j.jconrel.2017.08.023] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/20/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022]
Abstract
Nanomedicines have gained much attention for the delivery of small molecules or nucleic acids as treatment options for many diseases. However, the transfer from experimental systems to in vivo applications remains a challenge since it is difficult to assess their circulation behavior in the body at an early stage of drug discovery. Thus, innovative and improved concepts are urgently needed to overcome this issue and to close the gap between empiric nanoparticle design, in vitro assessment, and first in vivo experiments using rodent animal models. This study was focused on the zebrafish as a vertebrate screening model to assess the circulation in blood and extravasation behavior of nanoparticulate drug delivery systems in vivo. To validate this novel approach, monodisperse preparations of fluorescently labeled liposomes with similar size and zeta potential were injected into transgenic zebrafish lines expressing green fluorescent protein in their vasculature. Phosphatidylcholine-based lipids differed by fatty acid chain length and saturation. Circulation behavior and vascular distribution pattern were evaluated qualitatively and semi-quantitatively using image analysis. Liposomes composed of lipids with lower transition temperature (<28°C) as well as PEGylated liposomes showed longer circulation times and extravasation. In contrast, liposomes composed of lipids with transition temperatures>28°C bound to venous parts of the vasculature. This circulation patterns in the zebrafish model did correlate with published and experimental pharmacokinetic data from mice and rats. Our findings indicate that the zebrafish model is a useful vertebrate screening tool for nanoparticulate drug delivery systems to predict their in vivo circulation behavior with respect to systemic circulation time and exposure.
Collapse
Affiliation(s)
- Sandro Sieber
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Pascal Detampel
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Salome Siegfried
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
46
|
Chen CW, Syu WJ, Huang TC, Lee YC, Hsiao JK, Huang KY, Yu HP, Liao MY, Lai PS. Encapsulation of Au/Fe 3O 4 nanoparticles into a polymer nanoarchitecture with combined near infrared-triggered chemo-photothermal therapy based on intracellular secondary protein understanding. J Mater Chem B 2017; 5:5774-5782. [PMID: 32264211 DOI: 10.1039/c7tb00944e] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The combination of the functions of near infrared-triggered molecule release and chemo-photothermal therapy improved the therapeutic effect, but clarification of the cancer damage pathway in terms of protein molecule levels has yet to be well studied. In this study, we developed a polymer encapsulation synthesis of Au/Fe3O4@polymer nanoparticles as a Swiss army knife to integrate near infrared absorption, magnetism, and doxorubicin (DOX) loading ability into a single package. By exposing to near infrared absorption, the Au/Fe3O4@polymer nanoparticles possessed photothermal therapy, exhibiting anti-tumor growth suppression of HT-29 tumor-bearing nude mice with less body weight loss. To deeply understand the interactions between the drug-loaded nanocarriers and the protein structures of the treated cells, delivering therapeutic DOX agent combined with photothermal therapy with Au/Fe3O4@polymer nanostructures to cancer cells was investigated. Synchrotron-based FTIR imaging and confocal imaging showed direct observation of the efficient photo-chemotherapy impacting MCF7, MCF7/ADR, and HT-29 cells after the near infrared radiation-triggered DOX release. Our demonstration outlines how the cell destruction in the molecular mechanism was initiated by chemo-photothermal combination therapy after the translocation of DOX from the cytosol to the nuclei, leading to altered intracellular secondary proteins. For preclinical application of potential diagnosis to cancer cells, Au/Fe3O4@polymer nanoparticles performed integrated computed tomography/magnetic resonance imaging contrast enhancement and near infrared-triggered chemo-photothermal therapy.
Collapse
Affiliation(s)
- Ching-Wen Chen
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Summers HD, Rees P, Wang JTW, Al-Jamal KT. Spatially-resolved profiling of carbon nanotube uptake across cell lines. NANOSCALE 2017; 9:6800-6807. [PMID: 28489104 DOI: 10.1039/c7nr01561e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The internalisation and intra-cellular distribution of carbon nanotubes (CNT) has been quantitatively assessed using imaging flow cytometry. Spatial analysis of the bright field images indicates the presence of a small sub-population (5% of cells) in which the internalised CNTs are packed into pronounced clusters, visible as dark spots due to strong optical scattering by the nanotubes. The area of these spots can be used as a label-free metric of CNT dose and we assess the relative uptake of charge-neutral CNTs, over a 24 hours exposure period across four cell types: J774 mouse macrophage cells, A549 and Calu-6 human lung cancer cells, and MCF-7 human breast cells. The relative dose as indicated by the spot-area metric closely correlates to results using the same CNT preparation, conjugated to a FITC-label and shows pronounced uptake by the J774 cells leading to a mean dose that is >60% higher than for the other cell types. Spatial evaluation of dosing clusters is also used to quantify differences in uptake by J774 cells of CNTs with different surface functionalisation. While the percentage of CNT-cluster positive cells increases from 5% to 19% when switching from charge-neutral CNTs to poly-cationic, dendron functionalised CNTs, the single cell level analysis of internalised clusters indicates a lower dose per cell of poly-cationic CNTs relative to the charge-neutral CNTs. We concluded that there is dose homeostasis i.e., the population-averaged cellular dose of CNTs remained unchanged.
Collapse
Affiliation(s)
- H D Summers
- Centre for Nanohealth, College of Engineering, Swansea University, SA2 8PP, UK.
| | | | | | | |
Collapse
|
48
|
Ogunjimi AT, Melo SM, Vargas-Rechia CG, Emery FS, Lopez RF. Hydrophilic polymeric nanoparticles prepared from Delonix galactomannan with low cytotoxicity for ocular drug delivery. Carbohydr Polym 2017; 157:1065-1075. [DOI: 10.1016/j.carbpol.2016.10.076] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/20/2016] [Accepted: 10/24/2016] [Indexed: 11/29/2022]
|
49
|
Hinde E, Thammasiraphop K, Duong HTT, Yeow J, Karagoz B, Boyer C, Gooding JJ, Gaus K. Pair correlation microscopy reveals the role of nanoparticle shape in intracellular transport and site of drug release. NATURE NANOTECHNOLOGY 2017; 12:81-89. [PMID: 27618255 DOI: 10.1038/nnano.2016.160] [Citation(s) in RCA: 255] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 07/21/2016] [Indexed: 05/20/2023]
Abstract
Nanoparticle size, surface charge and material composition are known to affect the uptake of nanoparticles by cells. However, whether nanoparticle shape affects transport across various barriers inside the cell remains unclear. Here we used pair correlation microscopy to show that polymeric nanoparticles with different shapes but identical surface chemistries moved across the various cellular barriers at different rates, ultimately defining the site of drug release. We measured how micelles, vesicles, rods and worms entered the cell and whether they escaped from the endosomal system and had access to the nucleus via the nuclear pore complex. Rods and worms, but not micelles and vesicles, entered the nucleus by passive diffusion. Improving nuclear access, for example with a nuclear localization signal, resulted in more doxorubicin release inside the nucleus and correlated with greater cytotoxicity. Our results therefore demonstrate that drug delivery across the major cellular barrier, the nuclear envelope, is important for doxorubicin efficiency and can be achieved with appropriately shaped nanoparticles.
Collapse
Affiliation(s)
- Elizabeth Hinde
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney 2052, Australia
| | - Kitiphume Thammasiraphop
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney 2052, Australia
| | - Hien T T Duong
- Australian Centre for NanoMedicine, University of New South Wales, Sydney 2052, Australia
| | - Jonathan Yeow
- Australian Centre for NanoMedicine, University of New South Wales, Sydney 2052, Australia
- Centre for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney 2052, Australia
| | - Bunyamin Karagoz
- Australian Centre for NanoMedicine, University of New South Wales, Sydney 2052, Australia
- School of Chemistry, University of New South Wales, Sydney 2052, Australia
| | - Cyrille Boyer
- Australian Centre for NanoMedicine, University of New South Wales, Sydney 2052, Australia
- Centre for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney 2052, Australia
| | - J Justin Gooding
- Australian Centre for NanoMedicine, University of New South Wales, Sydney 2052, Australia
- School of Chemistry, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney 2052, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
50
|
Kaizuka Y, Ura T, Lyu S, Chao L, Henzie J, Nakao H. Cytosolic Transport of Nanoparticles through Pressurized Plasma Membranes for Molecular Delivery and Amplification of Intracellular Fluorescence. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:13534-13545. [PMID: 27993015 DOI: 10.1021/acs.langmuir.6b03412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Transporting nanoparticles into live cells is important for drug delivery and other related applications. We found that cells exposed to hypoosmotic pressures can internalize substantial quantities of gold nanoparticles. Importantly, these nanoparticles can circumvent normal intracellular traffic and be transported directly into the cytosol, without the need for surface functionalization. In contrast, nanoparticles endocytosed at physiological osmolality are segregated inside endocytic organelles and are not able to reach the cytosol. Cytosolic internalization was observed for nanoparticles of various sizes and materials, with minimal short- or long-term damage induced by the internalized particles. Thus, our strategy can be used as a delivery platform for a range of applications from therapeutics to medical imaging. As examples, we demonstrated rapid delivery of membrane-impermeable molecules to the cytosol by using nanoparticles as carriers and the use of nanoparticles assembled within the cytosol as plasmonic nanoantenna to enhance intracellular fluorescence. We propose a model for the mechanisms behind nanoparticle internalization through pressurized plasma membranes via the release of lateral pressures. Such characterizations may constitute a foundation for developing new technologies, including nanoparticle-based drug delivery.
Collapse
Affiliation(s)
- Yoshihisa Kaizuka
- National Institute for Materials Science , 1-2-1 Sengen, Tsukuba, Ibaraki 305-0047, Japan , and
| | - Tomoto Ura
- National Institute for Materials Science , 1-2-1 Sengen, Tsukuba, Ibaraki 305-0047, Japan , and
| | - Shaowei Lyu
- National Institute for Materials Science , 1-2-1 Sengen, Tsukuba, Ibaraki 305-0047, Japan , and
- Department of Chemical Engineering, National Taiwan University , Taipei 10617, Taiwan
| | - Ling Chao
- Department of Chemical Engineering, National Taiwan University , Taipei 10617, Taiwan
| | - Joel Henzie
- National Institute for Materials Science , 1-2-1 Sengen, Tsukuba, Ibaraki 305-0047, Japan , and
| | - Hidenobu Nakao
- National Institute for Materials Science , 1-2-1 Sengen, Tsukuba, Ibaraki 305-0047, Japan , and
| |
Collapse
|