1
|
Shivanna AT, Dash BS, Lu YJ, Lin WT, Chen JP. Magnetic lipid-poly(lactic-co-glycolic acid) nanoparticles conjugated with epidermal growth factor receptor antibody for dual-targeted delivery of CPT-11. Int J Pharm 2024; 667:124856. [PMID: 39461680 DOI: 10.1016/j.ijpharm.2024.124856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/01/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
To entrap sparingly water-soluble drugs like CPT-11 (irinotecan), the poly(lactic-co-glycolic acid) (PLGA) nanoparticle (NP) is highly favored due to its low cytotoxicity and approval for clinical use. On the other hand, entrapping hydrophobic oleic acid-coated iron oxide magnetic nanoparticles (OMNP) in PLGA NP can provide a nanovehicle for magnetically targeted drug delivery. Our goal in this study is to develop a new dual-targeted magnetic lipid-polymer NP for the delivery of CPT-11. We first co-entrap OMNP and CPT-11 in self-assembled lipid-PLGA NP to prepare OLNP@CPT-11. The OLNP@CPT-11 surface was modified with an epidermal growth factor receptor (EGFR) antibody Cetuximab (CET), which can actively target the overexpressed EGFR on the U87 glioblastoma cell surface. The OLNP-CET@CPT-11 enables dual targeting through both external magnetic guidance and CET-mediated active targeting. The NP was characterized for physicochemical properties using various analytical techniques. In vitro study confirms ligand-receptor interaction results in enhanced endocytosis of OLNP-CET@CPT-11 by U87 cells, which offers increased cytotoxicity and elevated cell apoptosis rates. Furthermore, magnetic guidance of OLNP-CET@CPT-11 to U87 cells can induce cell death exclusively in the magnetically targeted zone. The dual-targeted strategy also provides the best therapeutic efficacy against subcutaneously implanted U87 tumors in nude mice with intravenously delivered OLNP-CET@CPT-11.
Collapse
Affiliation(s)
- Anilkumar T Shivanna
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan
| | - Wei-Ting Lin
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan; Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan; Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
2
|
Khalili-Hezarjaribi H, Bahrami AR, Sh Saljooghi A, Matin MM. Modified mesoporous silica nanocarriers containing superparamagnetic iron oxide nanoparticle, 5-fluorouracil or oxaliplatin, and metformin as a radiosensitizer, significantly impact colorectal cancer radiation therapy. Int J Pharm 2024; 666:124838. [PMID: 39419365 DOI: 10.1016/j.ijpharm.2024.124838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/25/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024]
Abstract
This study investigates the anticancer effects of SPION-based silica nanoparticles carrying 5-fluorouracil (5-FU) or oxaliplatin (OX), and metformin (MET) on colorectal cancer cells. Nanocarriers were equipped with pH-responsive gold gatekeepers for controlled release, PEGylation for longer circulation, and folic acid (FA) for targeted delivery. The effects were evaluated by investigating cell viability, cellular uptake, flow cytometry, and clonogenic assay in vitro. The efficacy of the system was also tested in vivo on C57BL/6 mice bearing HT-29 tumors, and potential side effects were evaluated. Nanocarriers were synthesized with hydrodynamic diameters of 79.8 nm for 5-FU and 85.2 nm for OX; zeta potentials of -21 and -22 mV, respectively, and remained stable after 72 h. Encapsulation efficiencies were 85 % for 5-FU, 80 % for OX, and 83 % for MET, with loading capacities of 44 %, 38 %, and 41 %, respectively. Drug release in acidic buffer was 38.7 % for 5-FU, 32.8 % for OX, and 43.5 % for MET. MTT assay showed increased toxicity due to FA conjugation, while PEGylation reduced the hemolysis activity. Targeted nanocarriers demonstrated superior cellular uptake and tumor localization compared to non-targeted variants. The combination of 5-FU-MET and OX-MET nanocarriers with radiation therapy (RT) demonstrated the greatest effect on their antitumor activity, accompanied by minimal side effects indicating effective tumor targeting in vivo. MRI and CT imaging further supported these findings. This study underscores the synergistic impact of MET alongside RT on the inhibition of cancer cells and tumor growth for both targeted 5-FU and OX nanocarriers reflecting the significant radiosensitizing properties of MET.
Collapse
Affiliation(s)
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Sh Saljooghi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
3
|
Ning L, Zanella S, Tomov ML, Amoli MS, Jin L, Hwang B, Saadeh M, Chen H, Neelakantan S, Dasi LP, Avazmohammadi R, Mahmoudi M, Bauser‐Heaton HD, Serpooshan V. Targeted Rapamycin Delivery via Magnetic Nanoparticles to Address Stenosis in a 3D Bioprinted in Vitro Model of Pulmonary Veins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400476. [PMID: 38696618 PMCID: PMC11234432 DOI: 10.1002/advs.202400476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/09/2024] [Indexed: 05/04/2024]
Abstract
Vascular cell overgrowth and lumen size reduction in pulmonary vein stenosis (PVS) can result in elevated PV pressure, pulmonary hypertension, cardiac failure, and death. Administration of chemotherapies such as rapamycin have shown promise by inhibiting the vascular cell proliferation; yet clinical success is limited due to complications such as restenosis and off-target effects. The lack of in vitro models to recapitulate the complex pathophysiology of PVS has hindered the identification of disease mechanisms and therapies. This study integrated 3D bioprinting, functional nanoparticles, and perfusion bioreactors to develop a novel in vitro model of PVS. Bioprinted bifurcated PV constructs are seeded with endothelial cells (ECs) and perfused, demonstrating the formation of a uniform and viable endothelium. Computational modeling identified the bifurcation point at high risk of EC overgrowth. Application of an external magnetic field enabled targeting of the rapamycin-loaded superparamagnetic iron oxide nanoparticles at the bifurcation site, leading to a significant reduction in EC proliferation with no adverse side effects. These results establish a 3D bioprinted in vitro model to study PV homeostasis and diseases, offering the potential for increased throughput, tunability, and patient specificity, to test new or more effective therapies for PVS and other vascular diseases.
Collapse
Affiliation(s)
- Liqun Ning
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of Mechanical EngineeringCleveland State UniversityClevelandOH44115USA
| | - Stefano Zanella
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Martin L. Tomov
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Mehdi Salar Amoli
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Linqi Jin
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Boeun Hwang
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Maher Saadeh
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Huang Chen
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Sunder Neelakantan
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Lakshmi Prasad Dasi
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Reza Avazmohammadi
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
- J. Mike Walker ’66 Department of Mechanical EngineeringTexas A&M UniversityCollege StationTX77840USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health ProgramMichigan State UniversityEast LandingMI48824USA
| | - Holly D. Bauser‐Heaton
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
- Sibley Heart Center at Children's Healthcare of AtlantaAtlantaGA30322USA
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
| |
Collapse
|
4
|
Dezfuli AAZ, Abu-Elghait M, Salem SS. Recent Insights into Nanotechnology in Colorectal Cancer. Appl Biochem Biotechnol 2024; 196:4457-4471. [PMID: 37751009 DOI: 10.1007/s12010-023-04696-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/27/2023]
Abstract
Colorectal cancer (CRC) is the third cancer among the known causes of cancer that impact people. Although CRC drug options are imperfect, primary detection of CRC can play a key role in treating the disease and reducing mortality. Cancer tissues show many molecular markers that can be used as a new way to advance therapeutic methods. Nanotechnology includes a wide range of nanomaterials with high diagnostic and therapeutic power. Several nanomaterials and nanoformulations can be used to treat cancer, especially CRC. In this review, we discuss recent insights into nanotechnology in colorectal cancer.
Collapse
Affiliation(s)
- Aram Asareh Zadegan Dezfuli
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Mohammed Abu-Elghait
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Salem S Salem
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, Cairo, Egypt.
| |
Collapse
|
5
|
Shestovskaya MV, Luss AL, Bezborodova OA, Kulikov PP, Antufrieva DA, Plotnikova EA, Makarov VV, Yudin VS, Pankratov AA, Keskinov AA. Radiosensitizing effects of heparinized magnetic iron oxide nanoparticles in colon cancer. Biomed Pharmacother 2024; 175:116668. [PMID: 38701565 DOI: 10.1016/j.biopha.2024.116668] [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: 01/26/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/05/2024] Open
Abstract
The combination of radiation treatment and chemotherapy is currently the standard for management of cancer patients. However, safe doses do not often provide effective therapy, then pre-treated patients are forced to repeat treatment with often already increased tumor resistance to drugs and irradiation. One of the solutions we suggest is to improve primary course of radiation treatment via enhancing radiosensitivity of tumors by magnetic-guided iron oxide nanoparticles (magnetite). We obtained spherical heparinized iron oxide nanoparticles (hIONPs, ∼20 nm), characterized it by TEM, Infrared spectroscopy and DLS. Then hIONPs cytotoxicity was assessed for colon cancer cells (XTT assay) and cellular uptake of nanoparticles was analyzed with X-ray fluorescence. Combination of ionizing radiation (IR) and hIONPs in vitro caused an increase of G2/M arrest of cell cycle, mitotic errors and decrease in survival (compared with samples exposed to IR and hIONPs separately). The promising results were shown for magnetic-guided hIONPs in CT26-grafted BALB/C mice: the combination of intravenously administrated hIONPs and IR showed 20,8% T/C ratio (related to non-treated mice), while single radiation had no shown significant decrease in tumor growth (72,4%). Non-guided by magnets hIONPs with IR showed 57,9% of T/C. This indicates that ultra-small size and biocompatible molecule are not the key to successful nano-drug design, in each case, delivery technologies need to be improved when transferred to in vivo model.
Collapse
Affiliation(s)
- Maria V Shestovskaya
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Pogodinskaya st. 10/1, Moscow 119435, Russia.
| | - Anna L Luss
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Pogodinskaya st. 10/1, Moscow 119435, Russia
| | - Olga A Bezborodova
- P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinskiy p. 3, Moscow 125284, Russia
| | - Pavel P Kulikov
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Pogodinskaya st. 10/1, Moscow 119435, Russia
| | - Daria A Antufrieva
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Pogodinskaya st. 10/1, Moscow 119435, Russia
| | - Ekaterina A Plotnikova
- P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinskiy p. 3, Moscow 125284, Russia
| | - Valentin V Makarov
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Pogodinskaya st. 10/1, Moscow 119435, Russia
| | - Vladimir S Yudin
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Pogodinskaya st. 10/1, Moscow 119435, Russia
| | - Andrey A Pankratov
- P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinskiy p. 3, Moscow 125284, Russia
| | - Anton A Keskinov
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Pogodinskaya st. 10/1, Moscow 119435, Russia
| |
Collapse
|
6
|
Salvanou EA, Kolokithas-Ntoukas A, Prokopiou D, Theodosiou M, Efthimiadou E, Koźmiński P, Xanthopoulos S, Avgoustakis K, Bouziotis P. 177Lu-Labeled Iron Oxide Nanoparticles Functionalized with Doxorubicin and Bevacizumab as Nanobrachytherapy Agents against Breast Cancer. Molecules 2024; 29:1030. [PMID: 38474542 DOI: 10.3390/molecules29051030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
The use of conventional methods for the treatment of cancer, such as chemotherapy or radiotherapy, and approaches such as brachytherapy in conjunction with the unique properties of nanoparticles could enable the development of novel theranostic agents. The aim of our current study was to evaluate the potential of iron oxide nanoparticles, coated with alginic acid and polyethylene glycol, functionalized with the chemotherapeutic agent doxorubicin and the monoclonal antibody bevacizumab, to serve as a nanoradiopharmaceutical agent against breast cancer. Direct radiolabeling with the therapeutic isotope Lutetium-177 (177Lu) resulted in an additional therapeutic effect. Functionalization was accomplished at high percentages and radiolabeling was robust. The high cytotoxic effect of our radiolabeled and non-radiolabeled nanostructures was proven in vitro against five different breast cancer cell lines. The ex vivo biodistribution in tumor-bearing mice was investigated with three different ways of administration. The intratumoral administration of our functionalized radionanoconjugates showed high tumor accumulation and retention at the tumor site. Finally, our therapeutic efficacy study performed over a 50-day period against an aggressive triple-negative breast cancer cell line (4T1) demonstrated enhanced tumor growth retention, thus identifying the developed nanoparticles as a promising nanobrachytherapy agent against breast cancer.
Collapse
Affiliation(s)
- Evangelia-Alexandra Salvanou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research "Demokritos", 15341 Athens, Greece
| | | | - Danai Prokopiou
- Laboratory of Inorganic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece
| | - Maria Theodosiou
- Laboratory of Inorganic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece
| | - Eleni Efthimiadou
- Laboratory of Inorganic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece
| | - Przemysław Koźmiński
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16 Str., 03-195 Warsaw, Poland
| | - Stavros Xanthopoulos
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research "Demokritos", 15341 Athens, Greece
| | | | - Penelope Bouziotis
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research "Demokritos", 15341 Athens, Greece
| |
Collapse
|
7
|
Firouzi Amandi A, Bahmanyar Z, Dadashpour M, Lak M, Natami M, Döğüş Y, Alem M, Adeli OA. Fabrication of magnetic niosomal platform for delivery of resveratrol: potential anticancer activity against human pancreatic cancer Capan-1 cell. Cancer Cell Int 2024; 24:46. [PMID: 38287318 PMCID: PMC10826113 DOI: 10.1186/s12935-024-03219-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024] Open
Abstract
Recently, the presence of different nanoparticles (NPs) has developed targeting drug delivery in treatment of cancer cell. Targeted drug delivery systems using NPs have shown great promise in improving the efficacy of intracellular uptake as well as local concentration of therapeutics with minimizing side effects. The current study planned to synthesized resveratrol-loaded magnetic niosomes nanoparticles (RSV-MNIONPs) and evaluate their cytotoxicity activity in pancreatic cancer cells. For this aim, magnetic nanoparticles (MNPs) were synthesized and loaded into niosomes (NIOs) by the thin film hydration technique and then characterized via DLS, FT-IR, TEM, SEM and VSM techniques. Moreover, the cytotoxic activity of the RSV-MNIONPs on the Capan-1 cells line was assessed by the MTT test. The distribution number of RSV-MNIONPs was gained about 80 nm and 95 nm with surface charge of - 14.0 mV by SEM and TEM analysis, respectively. RSV loading efficacy in NIOs was about 85%, and the drug releases pattern displayed a sustained discharge with a maximum amount about 35% and 40%, within 4 h in pH = 7.4 and pH = 5.8, respectively. The cytotoxicity of the RSV-MNIONPs in the presence of an external magnetic field is higher than that of the RSV, indicating enhanced cellular uptake in their encapsulated states. Furthermore, RSV loaded MNNPs were found to induce more cell cycle arrest at the G0/G1 checkpoint than free RSV. Compared with RSV-treated cells, the mRNA expression levels of BAX, Bcl2, FAS, P 53, Cyclin D and hTERT, were significantly changed in cells treated with RSV loaded MNNPs. The niosomes NPs approaches have been widely used to attain higher solubility, improved bioavailability, enhanced stability, and control delivery of RSV. Our formulation displayed antitumor activity and can be considered an appropriate carrier with a great potential for future usage in cancer therapy.
Collapse
Affiliation(s)
- Akram Firouzi Amandi
- Department of Medical Immunology, Facultyof Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Bahmanyar
- School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dadashpour
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| | - Mehrnoosh Lak
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Natami
- Department of Urology, Shahid Mohammadi Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Yusuf Döğüş
- Department of Medical Biochemistry, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Mahsa Alem
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Omid Ali Adeli
- Department of Pathology, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
8
|
Shestovskaya MV, Luss AL, Bezborodova OA, Makarov VV, Keskinov AA. Iron Oxide Nanoparticles in Cancer Treatment: Cell Responses and the Potency to Improve Radiosensitivity. Pharmaceutics 2023; 15:2406. [PMID: 37896166 PMCID: PMC10610190 DOI: 10.3390/pharmaceutics15102406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The main concept of radiosensitization is making the tumor tissue more responsive to ionizing radiation, which leads to an increase in the potency of radiation therapy and allows for decreasing radiation dose and the concomitant side effects. Radiosensitization by metal oxide nanoparticles is widely discussed, but the range of mechanisms studied is not sufficiently codified and often does not reflect the ability of nanocarriers to have a specific impact on cells. This review is focused on the magnetic iron oxide nanoparticles while they occupied a special niche among the prospective radiosensitizers due to unique physicochemical characteristics and reactivity. We collected data about the possible molecular mechanisms underlying the radiosensitizing effects of iron oxide nanoparticles (IONPs) and the main approaches to increase their therapeutic efficacy by variable modifications.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| | - Anna L. Luss
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
- The Department of Technology of Chemical, Pharmaceutical and Cosmetic Products Mendeleev of University of Chemical Technology of Russia, Miusskaya sq. 9, Moscow 125047, Russia
| | - Olga A. Bezborodova
- P. Hertsen Moscow Oncology Research Institute of the National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinskiy p. 3, Moscow 125284, Russia;
| | - Valentin V. Makarov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| | - Anton A. Keskinov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| |
Collapse
|
9
|
Wang T, Wu C, Hu Y, Zhang Y, Ma J. Stimuli-responsive nanocarrier delivery systems for Pt-based antitumor complexes: a review. RSC Adv 2023; 13:16488-16511. [PMID: 37274408 PMCID: PMC10233443 DOI: 10.1039/d3ra00866e] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/30/2023] [Indexed: 06/06/2023] Open
Abstract
Platinum-based anticancer drugs play a crucial role in the clinical treatment of various cancers. However, the application of platinum-based drugs is heavily restricted by their severe toxicity and drug resistance/cross resistance. Various drug delivery systems have been developed to overcome these limitations of platinum-based chemotherapy. Stimuli-responsive nanocarrier drug delivery systems as one of the most promising strategies attract more attention. And huge progress in stimuli-responsive nanocarrier delivery systems of platinum-based drugs has been made. In these systems, a variety of triggers including endogenous and extracorporeal stimuli have been employed. Endogenous stimuli mainly include pH-, thermo-, enzyme- and redox-responsive nanocarriers. Extracorporeal stimuli include light-, magnetic field- and ultrasound responsive nanocarriers. In this review, we present the recent advances in stimuli-responsive drug delivery systems with different nanocarriers for improving the efficacy and reducing the side effects of platinum-based anticancer drugs.
Collapse
Affiliation(s)
- Tianshuai Wang
- Hubei Key Lab of Wudang Local Chinese Medicine Research, Hubei University of Medicine Shiyan 442000 Hubei China
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Chen Wu
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Yanggen Hu
- Hubei Key Lab of Wudang Local Chinese Medicine Research, Hubei University of Medicine Shiyan 442000 Hubei China
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Yan Zhang
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Junkai Ma
- Hubei Key Lab of Wudang Local Chinese Medicine Research, Hubei University of Medicine Shiyan 442000 Hubei China
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| |
Collapse
|
10
|
Dinter J, Friedrich RP, Yang H, Pilarsky C, Mangge H, Pöttler M, Janko C, Alexiou C, Lyer S. Mitoxantrone and Mitoxantrone-Loaded Iron Oxide Nanoparticles Induce Cell Death in Human Pancreatic Ductal Adenocarcinoma Cell Spheroids. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2906. [PMID: 37049199 PMCID: PMC10096321 DOI: 10.3390/ma16072906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 06/19/2023]
Abstract
Pancreatic ductal adenocarcinoma is a hard-to-treat, deadly malignancy. Traditional treatments, such as surgery, radiation and chemotherapy, unfortunately are still not able to significantly improve long-term survival. Three-dimensional (3D) cell cultures might be a platform to study new drug types in a highly reproducible, resource-saving model within a relevant pathophysiological cellular microenvironment. We used a 3D culture of human pancreatic ductal adenocarcinoma cell lines to investigate a potential new treatment approach using superparamagnetic iron oxide nanoparticles (SPIONs) as a drug delivery system for mitoxantrone (MTO), a chemotherapeutic agent. We established a PaCa DD183 cell line and generated PANC-1SMAD4 (-/-) cells by using the CRISPR-Cas9 system, differing in a prognostically relevant mutation in the TGF-β pathway. Afterwards, we formed spheroids using PaCa DD183, PANC-1 and PANC-1SMAD4 (-/-) cells, and analyzed the uptake and cytotoxic effect of free MTO and MTO-loaded SPIONs by microscopy and flow cytometry. MTO and SPION-MTO-induced cell death in all tumor spheroids in a dose-dependent manner. Interestingly, spheroids with a SMAD4 mutation showed an increased uptake of MTO and SPION-MTO, while at the same time being more resistant to the cytotoxic effects of the chemotherapeutic agents. MTO-loaded SPIONs, with their ability for magnetic drug targeting, could be a future approach for treating pancreatic ductal adenocarcinomas.
Collapse
Affiliation(s)
- Jonas Dinter
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Medical Faculty, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Ralf P. Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Hai Yang
- Department of Surgery, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Christian Pilarsky
- Department of Surgery, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnosis, Medical University of Graz, 8036 Graz, Austria
| | - Marina Pöttler
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Professorship for AI-Controlled Nanomaterials, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Professorship for AI-Controlled Nanomaterials, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
11
|
Curcio A, Perez JE, Prévéral S, Fromain A, Genevois C, Michel A, Van de Walle A, Lalatonne Y, Faivre D, Ménager C, Wilhelm C. The role of tumor model in magnetic targeting of magnetosomes and ultramagnetic liposomes. Sci Rep 2023; 13:2278. [PMID: 36755030 PMCID: PMC9908874 DOI: 10.1038/s41598-023-28914-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
The combined passive and active targeting of tumoral tissue remains an active and relevant cancer research field. Here, we exploit the properties of two highly magnetic nanomaterials, magnetosomes and ultramagnetic liposomes, in order to magnetically target prostate adenocarcinoma tumors, implanted orthotopically or subcutaneously, to take into account the role of tumor vascularization in the targeting efficiency. Analysis of organ biodistribution in vivo revealed that, for all conditions, both nanomaterials accumulate mostly in the liver and spleen, with an overall low tumor retention. However, both nanomaterials were more readily identified in orthotopic tumors, reflecting their higher tumor vascularization. Additionally, a 2- and 3-fold increase in nanomaterial accumulation was achieved with magnetic targeting. In summary, ultramagnetic nanomaterials show promise mostly in the targeting of highly-vascularized orthotopic murine tumor models.
Collapse
Affiliation(s)
- Alberto Curcio
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005, Paris, France
| | - Jose Efrain Perez
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005, Paris, France
| | - Sandra Prévéral
- Aix-Marseille University (AMU), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), UMR7265 Institute of Biosciences and Biotechnologies of Aix-Marseille (BIAM), 13108, Saint-Paul-lez-Durance, France
| | - Alexandre Fromain
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005, Paris, France
| | - Coralie Genevois
- TBM Core, UAR 3427, INSERM US 005, University of Bordeaux, 33000, Bordeaux, France
| | - Aude Michel
- Laboratoire Physicochimie des Électrolytes et Nanosystèmes Interfaciaux, CNRS, Sorbonne Université, Phenix, 75005, Paris, France
| | - Aurore Van de Walle
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005, Paris, France
| | - Yoann Lalatonne
- Université Sorbonne Paris Nord, Université Paris Cité, Laboratory for Vascular Translational Science, LVTS, INSERM, UMR 1148, Bobigny, F-93017, France
- Département de Biophysique et de Médecine Nucléaire, Assistance Publique-Hôpitaux de Paris, Hôpital Avicenne F- 93009, Bobigny, France
| | - Damien Faivre
- Aix-Marseille University (AMU), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), UMR7265 Institute of Biosciences and Biotechnologies of Aix-Marseille (BIAM), 13108, Saint-Paul-lez-Durance, France
| | - Christine Ménager
- Laboratoire Physicochimie des Électrolytes et Nanosystèmes Interfaciaux, CNRS, Sorbonne Université, Phenix, 75005, Paris, France
| | - Claire Wilhelm
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005, Paris, France.
| |
Collapse
|
12
|
Khizar S, Elkalla E, Zine N, Jaffrezic-Renault N, Errachid A, Elaissari A. Magnetic nanoparticles: multifunctional tool for cancer therapy. Expert Opin Drug Deliv 2023; 20:189-204. [PMID: 36608938 DOI: 10.1080/17425247.2023.2166484] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Cancer has one of the highest mortality rates globally. The traditional therapies used to treat cancer have harmful adverse effects. Considering these facts, researchers have explored new therapeutic possibilities with enhanced benefits. Nanoparticle development for cancer detection, in addition to therapy, has shown substantial progress over the past few years. AREA COVERED Herein, the latest research regarding cancer treatment employing magnetic nanoparticles (MNPs) in chemo-, immuno-, gene-, and radiotherapy along with hyperthermia is summarized, in addition to their physio-chemical features, advantages, and limitations for clinical translation have also been discussed. EXPERT OPINION MNPs are being extensively investigated and developed into effective modules for cancer therapy. They are highly functional tools aimed at cancer therapy owing to their excellent superparamagnetic, chemical, biocompatible, physical, and biodegradable properties.
Collapse
Affiliation(s)
- Sumera Khizar
- Univ Lyon, University Cla-ude Bernard Lyon-1, CNRS, ISA-UMR 5280, Lyon, France
| | - Eslam Elkalla
- Univ Lyon, University Cla-ude Bernard Lyon-1, CNRS, ISA-UMR 5280, Lyon, France
| | - Nadia Zine
- Univ Lyon, University Cla-ude Bernard Lyon-1, CNRS, ISA-UMR 5280, Lyon, France
| | | | - Abdelhamid Errachid
- Univ Lyon, University Cla-ude Bernard Lyon-1, CNRS, ISA-UMR 5280, Lyon, France
| | | |
Collapse
|
13
|
Zhu Y, Yang D, Guo T, Lin M. Use of S2.2/DOX Magnetic Nanoliposomes in MR Molecule Imaging and Targeted Thermochemotherapy for Breast Cancer In Vitro. Technol Cancer Res Treat 2023; 22:15330338231194498. [PMID: 37563954 PMCID: PMC10422896 DOI: 10.1177/15330338231194498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE To prepare S2.2/DOX magnetic nanoliposomes by combining the potential benefits of MNPs in MRI and the targeted performance of nano-drugs as an innovative method for integrated diagnosis and treatment of breast cancer (BC). METHODS We created a S2.2-PEG-MZF/DOX molecular probe by using a lipid material to encapsulate PEG-MZF-NPs and doxorubicin (DOX), and a S2.2 aptamer to target MUC1 to conjugate with PEG-MZF/DOX nanoliposomes. The potential of probe for cell-specific targeting and magnetic resonance (MR) molecular imaging was evaluated by MR scanner and Prussian blue staining. Additionally, we explored the feasibility by using nanoliposome magnetic induction heating to interfere with MCF-7 (MUC1+) BC cells under the influence of an alternating magnetic field (AMF). RESULTS PEG-MZF-NPs were biologically safe. The T2 relaxation rate of PEG-MZF-NPs was found to inhibit T2 signal in a concentration-dependent manner, and the T2 signal of the S2.2-PEG-MZF molecular probe in MCF-7 cells was significantly lower than that in PEG-MZF-NPs group. Moreover, the T2 signal reduction was more pronounced in MCF-7 cells than in the hepatoma cell line HepG2 (MUC1-), suggesting a strong MRI potential of the S2.2-PEG-MZF molecular probe. The S2.2-PEG-MZF/DOX nanoliposome was able to achieve the desired temperature range for tumor hyperthermia (42-44 °C) in vitro. The S2.2-PEG-MZF/DOX nanoliposome accompanied by magnetic fluid hyperthermia (MFH) could inhibit proliferation and invasion and induce apoptosis of MCF-7 cells. The effects of this approach were significantly higher than those observed in the other groups. CONCLUSION We successfully developed a novel technique for BC diagnosis and treatment using thermochemotherapy under the guidance of MR molecular imaging. This approach holds great potential for improving the management of this devastating disease in the future.
Collapse
Affiliation(s)
- Yinxing Zhu
- Taizhou School of Clinical Medicine, Nanjing Medical University, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Dazhuang Yang
- Imaging Department, General Hospital of Xuzhou Mining Group, Xuzhou, Jiangsu, China
| | - Ting Guo
- Institute of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Mei Lin
- Clinical Laboratory, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| |
Collapse
|
14
|
Formation of miRNA Nanoprobes-Conjugation Approaches Leading to the Functionalization. Molecules 2022; 27:molecules27238428. [PMID: 36500520 PMCID: PMC9739806 DOI: 10.3390/molecules27238428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Recently, microRNAs (miRNA) captured the interest as novel diagnostic and prognostic biomarkers, with their potential for early indication of numerous pathologies. Since miRNA is a short, non-coding RNA sequence, the sensitivity and selectivity of their detection remain a cornerstone of scientific research. As such, methods based on nanomaterials have emerged in hopes of developing fast and facile approaches. At the core of the detection method based on nanotechnology lie nanoprobes and other functionalized nanomaterials. Since miRNA sensing and detection are generally rooted in the capture of target miRNA with the complementary sequence of oligonucleotides, the sequence needs to be attached to the nanomaterial with a specific conjugation strategy. As each nanomaterial has its unique properties, and each conjugation approach presents its drawbacks and advantages, this review offers a condensed overview of the conjugation approaches in nanomaterial-based miRNA sensing. Starting with a brief recapitulation of specific properties and characteristics of nanomaterials that can be used as a substrate, the focus is then centered on covalent and non-covalent bonding chemistry, leading to the functionalization of the nanomaterials, which are the most commonly used in miRNA sensing methods.
Collapse
|
15
|
Sikorski J, Drozd M, Matczuk M. Red Flags and Adversities on the Way to the Robust CE-ICP-MS/MS Quantitative Monitoring of Self-Synthesized Magnetic Iron Oxide(II, III)-Based Nanoparticle Interactions with Human Serum Proteins. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238442. [PMID: 36500533 PMCID: PMC9739417 DOI: 10.3390/molecules27238442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
The growing interest in superparamagnetic iron oxide nanoparticles (SPIONs) as potential theranostic agents is related to their unique properties and the broad range of possibilities for their surface functionalization. However, despite the rapidly expanding list of novel SPIONs with potential biomedical applications, there is still a lack of methodologies that would allow in-depth investigation of the interactions of those nanoparticles with biological compounds in human serum. Herein, we present attempts to employ capillary electrophoresis-inductively coupled plasma tandem mass spectrometry (CE-ICP-MS/MS) for this purpose and various obstacles and limitations noticed during the research. The CE and ICP-MS/MS parameters were optimized, and the developed method was used to study the interactions of two different proteins (albumin and transferrin) with various synthesized SPIONs. While the satisfactory resolution between proteins was obtained and the method was applied to examine individual reagents, it was revealed that the conjugates formed during the incubation of the proteins with SPIONs were not stable under the conditions of electrophoretic separation.
Collapse
Affiliation(s)
- Jacek Sikorski
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - Marcin Drozd
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
- Centre for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poleczki St. 19, 02-822 Warsaw, Poland
| | - Magdalena Matczuk
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
- Correspondence: ; Tel.: +48-(22)-2347719
| |
Collapse
|
16
|
Alexiou C. Emerging applications of magnetic nanoparticles in medicine – A personal perspective. Biochem Biophys Res Commun 2022; 633:52-54. [DOI: 10.1016/j.bbrc.2022.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/05/2022] [Accepted: 09/05/2022] [Indexed: 11/06/2022]
|
17
|
Mdlovu NV, Lin KS, Weng MT, Lin YS, Liu SY. Preparation and in-vitro/in-vivo evaluation of doxorubicin-loaded magnetic SBA-15 nanocomposites from rice husk for enhancing therapeutic efficacy. Colloids Surf B Biointerfaces 2022; 220:112923. [DOI: 10.1016/j.colsurfb.2022.112923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/27/2022]
|
18
|
Smith L, Kuncic Z, Byrne HL, Waddington D. Nanoparticles for MRI-guided radiation therapy: a review. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00145-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AbstractThe development of nanoparticle agents for MRI-guided radiotherapy is growing at an increasing pace, with clinical trials now underway and many pre-clinical evaluation studies ongoing. Gadolinium and iron-oxide-based nanoparticles remain the most clinically advanced nanoparticles to date, although several promising candidates are currently under varying stages of development. Goals of current and future generation nanoparticle-based contrast agents for MRI-guided radiotherapy include achieving positive signal contrast on T1-weighted MRI scans, local radiation enhancement at clinically relevant concentrations and, where applicable, avoidance of uptake by the reticuloendothelial system. Exploiting the enhanced permeability and retention effect or the use of active targeting ligands on nanoparticle surfaces is utilised to promote tumour uptake. This review outlines the current status of promising nanoparticle agents for MRI-guided radiation therapy, including several platforms currently undergoing clinical evaluation or at various stages of the pre-clinical development process. Challenges facing nanoparticle agents and possible avenues for current and future development are discussed.
Collapse
|
19
|
Combination of Nanovectorized siRNA Directed against Survivin with Doxorubicin for Efficient Anti-Cancer Activity in HER2+ Breast Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14112537. [PMID: 36432729 PMCID: PMC9696063 DOI: 10.3390/pharmaceutics14112537] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/04/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
According to Globocan 2020, breast cancer is considered one of the most common cancers affecting women and is one of the leading causes of death in over 100 countries. The available classical treatment options do not always give satisfactory outcomes, and some patients develop resistance to these treatments. This study aims to investigate the combination of nanovectorized siRNA directed against anti-apoptotic protein Survivin (siSurvivin) by targeted stealth magnetic siRNA nanovectors (TS-MSN), designed in our lab, with Doxorubicin (DOX), as an option for HER2+ breast cancer treatment. The hypothesis is that the pretreatment of the HER2+ breast cancer cell line SK-BR-3 with siSurvivin will induce apoptosis in the cancer cells and enhance the therapeutic efficacy of DOX, allowing a dose reduction of DOX and hence a reduction of potential side effects. TS-MSN are based on superparamagnetic iron oxide nanoparticles (SPIONs) covalently coupled with a fluorophore sulfocyanine-5 and polyethylene glycol 5000 (PEG5000) and functionalized with single-chain variable fragments (scFv) of an antibody targeting the HER2 membrane receptor. These covalently functionalized SPIONs are then complexed via electrostatic interactions with therapeutic siRNA and the cationic polymers, chitosan, and poly-L-arginine. TS-MSNsiSurvivin had an average size of 144 ± 30 nm, a PDI of 0.3, and a slightly positive zeta potential value of 10.56 ± 05.70 mV. The agarose gel electrophoresis assay confirmed that the siRNA is well-complexed into TS-MSN without leakage, as no free siRNA was detected. Moreover, siRNA in TS-MSN was protected from RNAse A degradation for up to 6 h at 37 °C. Formulations of TS-MSN with siSurvivin demonstrated in vitro gene knockdown up to 89% in the HER2+ breast cancer cell line SK-BR-3. Furthermore, qRT-PCR confirmed a significant Survivin mRNA relative expression inhibition (about 50%) compared to control siRNA or untreated cells. A combination protocol was evaluated between TS-MSN and Doxorubicin (DOX) for the first time. Therefore, SK-BR-3 cells were pretreated with TS-MSN formulated with siSurvivin at 50 nM for 24 h alone, before a DOX treatment at a concentration of 0.5 µM (corresponding to the IC50) was added for 48 h. The MTT cytotoxicity tests, performed after 72 h of treatment, revealed that the combination had a significant synergistic cytotoxic effect on SK-BR-3 cells compared to monotherapies or untreated cells. We confirmed that pretreatment of cells with siSurvivin potentializes the cytotoxic effect of DOX as an alternative approach for treating HER2+ breast cancer. In conclusion, a combination of anti-Survivin siRNA and DOX would be a good alternative in HER2+ breast cancer therapy.
Collapse
|
20
|
Li J, Zhang X, Mo Y, Huang T, Rao H, Tan Z, Huang L, Zeng D, Jiang C, Zhong Y, Cai Y, Liang B, Wu J. Urokinase-loaded cyclic RGD-decorated liposome targeted therapy for in-situ thrombus of pulmonary arteriole of pulmonary hypertension. Front Bioeng Biotechnol 2022; 10:1038829. [PMID: 36324896 PMCID: PMC9618629 DOI: 10.3389/fbioe.2022.1038829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/26/2022] [Indexed: 09/07/2024] Open
Abstract
Backgroud: In-situ thrombosis is a significant pathophysiological basis for the development of pulmonary hypertension (PH). However, thrombolytic therapy for in-situ thrombus in PH was often hampered by the apparent side effects and the low bioavailability of common thrombolytic medications. Nanoscale cyclic RGD (cRGD)-decorated liposomes have received much attention thanks to their thrombus-targeting and biodegradability properties. As a result, we synthesized urokinase-loaded cRGD-decorated liposome (UK-cRGD-Liposome) for therapy of in-situ thrombosis as an exploration of pulmonary hypertensive novel therapeutic approaches. Purpose: To evaluate the utilize of UK-cRGD-Liposome for targeted thrombolysis of in-situ thrombus in PH and to explore the potential mechanisms of in-situ thrombus involved in the development of PH. Methods: UK-cRGD-Liposome nanoscale drug delivery system was prepared using combined methods of thin-film hydration and sonication. Induced PH via subcutaneous injection of monocrotaline (MCT). Fibrin staining (modified MSB method) was applied to detect the number of vessels within-situ thrombi in PH. Echocardiography, hematoxylin-eosin (H & E) staining, and Masson's trichrome staining were used to analyze right ventricular (RV) function, pulmonary vascular remodeling, as well as RV remodeling. Results: The number of vessels with in-situ thrombi revealed that UK-cRGD-Liposome could actively target urokinase to in-situ thrombi and release its payload in a controlled manner in the in vivo environment, thereby enhancing the thrombolytic effect of urokinase. Pulmonary artery hemodynamics and echocardiography indicated a dramatical decrease in pulmonary artery pressure and a significant improvement in RV function post targeted thrombolytic therapy. Moreover, pulmonary vascular remodeling and RV remodeling were significantly restricted post targeted thrombolytic therapy. Conclusion: UK-cRGD-Liposome can restrict the progression of PH and improve RV function by targeting the dissolution of pulmonary hypertensive in-situ thrombi, which may provide promising therapeutic approaches for PH.
Collapse
Affiliation(s)
- Jingtao Li
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaofeng Zhang
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yingying Mo
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tongtong Huang
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huaqing Rao
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhenyuan Tan
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Liuliu Huang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Decai Zeng
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chunlan Jiang
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yanfen Zhong
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yongzhi Cai
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Binbin Liang
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ji Wu
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
21
|
Metal nanoparticles: biomedical applications and their molecular mechanisms of toxicity. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-022-02351-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
22
|
Naseri E, Ahmadi A. A review on wound dressings: Antimicrobial agents, biomaterials, fabrication techniques, and stimuli-responsive drug release. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111293] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
23
|
Sikorski J, Matczuk M, Kamińska A, Kruszewska J, Trzaskowski M, Timerbaev AR, Jarosz M. Protein-Mediated Transformations of Superparamagnetic Nanoparticles Evidenced by Single-Particle Inductively Coupled Plasma Tandem Mass Spectrometry: A Disaggregation Phenomenon. Int J Mol Sci 2022; 23:ijms23031088. [PMID: 35163012 PMCID: PMC8835430 DOI: 10.3390/ijms23031088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Progress toward translating superparamagnetic iron oxide nanoparticles (SPIONs) with specific diagnostic and therapeutic properties for clinical applications depends on developing and implementing appropriate methodologies that would allow in-depth characterizations of their behavior in a real biological environment. Herein, we report a versatile approach for studying interactions between SPIONs and proteins using single-particle inductively coupled plasma tandem mass spectrometry. By monitoring the changes in the size distribution upon exposure to human serum, the formation of stable protein corona is revealed, accompanied by particle disaggregation.
Collapse
Affiliation(s)
- Jacek Sikorski
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland; (J.S.); (A.K.); (J.K.); (M.J.)
| | - Magdalena Matczuk
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland; (J.S.); (A.K.); (J.K.); (M.J.)
- Correspondence: ; Tel.: +48-(22)-2347719
| | - Agnieszka Kamińska
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland; (J.S.); (A.K.); (J.K.); (M.J.)
| | - Joanna Kruszewska
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland; (J.S.); (A.K.); (J.K.); (M.J.)
| | - Maciej Trzaskowski
- Centre for Advanced Materials and Technologies CEZAMAT PW, Warsaw University of Technology, Poleczki St. 19, 02-822 Warsaw, Poland;
| | - Andrei R. Timerbaev
- Vernadsky Institute of Geochemistry and Analytical Chemistry, Kosygin St. 19, 119991 Moscow, Russia;
| | - Maciej Jarosz
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland; (J.S.); (A.K.); (J.K.); (M.J.)
| |
Collapse
|
24
|
Liu L, Kshirsagar PG, Gautam SK, Gulati M, Wafa EI, Christiansen JC, White BM, Mallapragada SK, Wannemuehler MJ, Kumar S, Solheim JC, Batra SK, Salem AK, Narasimhan B, Jain M. Nanocarriers for pancreatic cancer imaging, treatments, and immunotherapies. Theranostics 2022; 12:1030-1060. [PMID: 35154473 PMCID: PMC8771545 DOI: 10.7150/thno.64805] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 12/03/2021] [Indexed: 01/28/2023] Open
Abstract
Pancreatic tumors are highly desmoplastic and immunosuppressive. Delivery and distribution of drugs within pancreatic tumors are compromised due to intrinsic physical and biochemical stresses that lead to increased interstitial fluid pressure, vascular compression, and hypoxia. Immunotherapy-based approaches, including therapeutic vaccines, immune checkpoint inhibition, CAR-T cell therapy, and adoptive T cell therapies, are challenged by an immunosuppressive tumor microenvironment. Together, extensive fibrosis and immunosuppression present major challenges to developing treatments for pancreatic cancer. In this context, nanoparticles have been extensively studied as delivery platforms and adjuvants for cancer and other disease therapies. Recent advances in nanotechnology have led to the development of multiple nanocarrier-based formulations that not only improve drug delivery but also enhance immunotherapy-based approaches for pancreatic cancer. This review discusses and critically analyzes the novel nanoscale strategies that have been used for drug delivery and immunomodulation to improve treatment efficacy, including newly emerging immunotherapy-based approaches. This review also presents important perspectives on future research directions that will guide the rational design of novel and robust nanoscale platforms to treat pancreatic tumors, particularly with respect to targeted therapies and immunotherapies. These insights will inform the next generation of clinical treatments to help patients manage this debilitating disease and enhance survival rates.
Collapse
Affiliation(s)
- Luman Liu
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA
| | - Prakash G. Kshirsagar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE
| | - Shailendra K. Gautam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE
| | - Mansi Gulati
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE
| | - Emad I. Wafa
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA
| | - John C. Christiansen
- Department of Veterinary Microbiology & Preventive Medicine, Iowa State University, Ames, IA
| | - Brianna M. White
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA
| | - Surya K. Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA
- Nanovaccine Institute, Iowa State University, Ames, IA
| | - Michael J. Wannemuehler
- Department of Veterinary Microbiology & Preventive Medicine, Iowa State University, Ames, IA
- Nanovaccine Institute, Iowa State University, Ames, IA
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE
| | - Joyce C. Solheim
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE
- Nanovaccine Institute, Iowa State University, Ames, IA
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha NE
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE
- Nanovaccine Institute, Iowa State University, Ames, IA
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha NE
| | - Aliasger K. Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA
- Nanovaccine Institute, Iowa State University, Ames, IA
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA
- Nanovaccine Institute, Iowa State University, Ames, IA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE
- Nanovaccine Institute, Iowa State University, Ames, IA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha NE
| |
Collapse
|
25
|
Anticancer Activity of 5-Fluorouracil-Loaded Nanoemulsions Containing Fe3O4/Au Core-Shell Nanoparticles. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.131075] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
26
|
Magnetic Nanoparticles Used in Oncology. MATERIALS 2021; 14:ma14205948. [PMID: 34683540 PMCID: PMC8539633 DOI: 10.3390/ma14205948] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/25/2021] [Accepted: 10/08/2021] [Indexed: 12/16/2022]
Abstract
Recently, magnetic nanoparticles (MNPs) have more and more often been used in experimental studies on cancer treatments, which have become one of the biggest challenges in medical research. The main goal of this research is to treat and to cure advanced or metastatic cancer with minimal side effects through nanotechnology. Drug delivery approaches take into account the fact that MNPs can be bonded to chemotherapeutical drugs, nucleic acids, synthetized antibodies or radionuclide substances. MNPs can be guided, and different treatment therapies can be applied, under the influence of an external magnetic field. This paper reviews the main MNPs’ synthesis methods, functionalization with different materials and highlight the applications in cancer therapy. In this review, we describe cancer cell monitorization based on different types of magnetic nanoparticles, chemotherapy, immunotherapy, magnetic hyperthermia, gene therapy and ferroptosis. Examples of applied treatments on murine models or humans are analyzed, and glioblastoma cancer therapy is detailed in the review. MNPs have an important contribution to diagnostics, investigation, and therapy in the so called theranostics domain. The main conclusion of this paper is that MNPs are very useful in different cancer therapies, with limited side effects, and they can increase the life expectancy of patients with cancer drug resistance.
Collapse
|
27
|
Phenotypic Switching of B16F10 Melanoma Cells as a Stress Adaptation Response to Fe3O4/Salicylic Acid Nanoparticle Therapy. Pharmaceuticals (Basel) 2021; 14:ph14101007. [PMID: 34681232 PMCID: PMC8537856 DOI: 10.3390/ph14101007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 02/06/2023] Open
Abstract
Melanoma is a melanocyte-derived skin cancer that has a high heterogeneity due to its phenotypic plasticity, a trait that may explain its ability to survive in the case of physical or molecular aggression and to develop resistance to therapy. Therefore, the therapy modulation of phenotypic switching in combination with other treatment modalities could become a common approach in any future therapeutic strategy. In this paper, we used the syngeneic model of B16F10 melanoma implanted in C57BL/6 mice to evaluate the phenotypic changes in melanoma induced by therapy with iron oxide nanoparticles functionalized with salicylic acid (SaIONs). The results of this study showed that the oral administration of the SaIONs aqueous dispersion was followed by phenotypic switching to highly pigmented cells in B16F10 melanoma through a cytotoxicity-induced cell selection mechanism. The hyperpigmentation of melanoma cells by the intra- or extracellular accumulation of melanic pigment deposits was another consequence of the SaIONs therapy. Additional studies are needed to assess the reversibility of SaIONs-induced phenotypic switching and the impact of tumor hyperpigmentation on B16F10 melanoma’s progression and metastasis abilities.
Collapse
|
28
|
Libring S, Enríquez Á, Lee H, Solorio L. In Vitro Magnetic Techniques for Investigating Cancer Progression. Cancers (Basel) 2021; 13:4440. [PMID: 34503250 PMCID: PMC8430481 DOI: 10.3390/cancers13174440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/28/2021] [Accepted: 08/29/2021] [Indexed: 12/24/2022] Open
Abstract
Worldwide, there are currently around 18.1 million new cancer cases and 9.6 million cancer deaths yearly. Although cancer diagnosis and treatment has improved greatly in the past several decades, a complete understanding of the complex interactions between cancer cells and the tumor microenvironment during primary tumor growth and metastatic expansion is still lacking. Several aspects of the metastatic cascade require in vitro investigation. This is because in vitro work allows for a reduced number of variables and an ability to gather real-time data of cell responses to precise stimuli, decoupling the complex environment surrounding in vivo experimentation. Breakthroughs in our understanding of cancer biology and mechanics through in vitro assays can lead to better-designed ex vivo precision medicine platforms and clinical therapeutics. Multiple techniques have been developed to imitate cancer cells in their primary or metastatic environments, such as spheroids in suspension, microfluidic systems, 3D bioprinting, and hydrogel embedding. Recently, magnetic-based in vitro platforms have been developed to improve the reproducibility of the cell geometries created, precisely move magnetized cell aggregates or fabricated scaffolding, and incorporate static or dynamic loading into the cell or its culture environment. Here, we will review the latest magnetic techniques utilized in these in vitro environments to improve our understanding of cancer cell interactions throughout the various stages of the metastatic cascade.
Collapse
Affiliation(s)
- Sarah Libring
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (Á.E.)
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
| | - Ángel Enríquez
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (Á.E.)
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
- Center for Implantable Devices, Purdue University, West Lafayette, IN 47907, USA
| | - Hyowon Lee
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (Á.E.)
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
- Center for Implantable Devices, Purdue University, West Lafayette, IN 47907, USA
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (Á.E.)
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
29
|
Suciu M, Mirescu C, Crăciunescu I, Macavei SG, Leoștean C, Ştefan R, Olar LE, Tripon SC, Ciorîță A, Barbu-Tudoran L. In Vivo Distribution of Poly(ethylene glycol) Functionalized Iron Oxide Nanoclusters: An Ultrastructural Study. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2184. [PMID: 34578500 PMCID: PMC8469409 DOI: 10.3390/nano11092184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 01/10/2023]
Abstract
The in vivo distribution of 50 nm clusters of polyethylene glycol-conjugated superparamagnetic iron oxide nanoparticles (SPIONs-PEG) was conducted in this study. SPIONs-PEG were synthesized de novo, and their structure and paramagnetic behaviors were analyzed by specific methods (TEM, DLS, XRD, VSM). Wistar rats were treated with 10 mg Fe/kg body weight SPIONs-PEG and their organs and blood were examined at two intervals for short-term (15, 30, 60, 180 min) and long-term (6, 12, 24 h) exposure evaluation. Most exposed organs were investigated through light and transmission electron microscopy, and blood and urine samples were examined through fluorescence spectrophotometry. SPIONs-PEG clusters entered the bloodstream after intraperitoneal and intravenous administrations and ended up in the urine, with the highest clearance at 12 h. The skin and spleen were within normal histological parameters, while the liver, kidney, brain, and lungs showed signs of transient local anoxia or other transient pathological affections. This study shows that once internalized, the synthesized SPIONs-PEG disperse well through the bloodstream with minor to nil induced tissue damage, are biocompatible, have good clearance, and are suited for biomedical applications.
Collapse
Affiliation(s)
- Maria Suciu
- Electron Microscopy Centre, Faculty of Biology and Geology, Babeș-Bolyai University, 44 Republicii St., 400015 Cluj-Napoca, Romania; (M.S.); (C.M.); (S.-C.T.)
- Integrated Electron Microscopy Laboratory, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat St., 400293 Cluj-Napoca, Romania
| | - Claudiu Mirescu
- Electron Microscopy Centre, Faculty of Biology and Geology, Babeș-Bolyai University, 44 Republicii St., 400015 Cluj-Napoca, Romania; (M.S.); (C.M.); (S.-C.T.)
| | - Izabell Crăciunescu
- Physics of Nanostructured Systems Department, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat, 400293 Cluj-Napoca, Romania; (I.C.); (S.G.M.); (C.L.)
| | - Sergiu Gabriel Macavei
- Physics of Nanostructured Systems Department, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat, 400293 Cluj-Napoca, Romania; (I.C.); (S.G.M.); (C.L.)
| | - Cristian Leoștean
- Physics of Nanostructured Systems Department, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat, 400293 Cluj-Napoca, Romania; (I.C.); (S.G.M.); (C.L.)
| | - Rǎzvan Ştefan
- Research Centre for Biophysics, Life Sciences Institute, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 3-5 Manastur St., 400372 Cluj-Napoca, Romania; (R.Ş.); (L.E.O.)
| | - Loredana E. Olar
- Research Centre for Biophysics, Life Sciences Institute, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 3-5 Manastur St., 400372 Cluj-Napoca, Romania; (R.Ş.); (L.E.O.)
| | - Septimiu-Cassian Tripon
- Electron Microscopy Centre, Faculty of Biology and Geology, Babeș-Bolyai University, 44 Republicii St., 400015 Cluj-Napoca, Romania; (M.S.); (C.M.); (S.-C.T.)
- Integrated Electron Microscopy Laboratory, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat St., 400293 Cluj-Napoca, Romania
| | - Alexandra Ciorîță
- Electron Microscopy Centre, Faculty of Biology and Geology, Babeș-Bolyai University, 44 Republicii St., 400015 Cluj-Napoca, Romania; (M.S.); (C.M.); (S.-C.T.)
- Integrated Electron Microscopy Laboratory, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat St., 400293 Cluj-Napoca, Romania
| | - Lucian Barbu-Tudoran
- Electron Microscopy Centre, Faculty of Biology and Geology, Babeș-Bolyai University, 44 Republicii St., 400015 Cluj-Napoca, Romania; (M.S.); (C.M.); (S.-C.T.)
- Integrated Electron Microscopy Laboratory, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat St., 400293 Cluj-Napoca, Romania
| |
Collapse
|
30
|
Mol B, Beeran AE, Jayaram PS, Prakash P, Jayasree RS, Thomas S, Chakrapani B, Anantharaman MR, Bushiri MJ. Radio frequency plasma assisted surface modification of Fe 3O 4 nanoparticles using polyaniline/polypyrrole for bioimaging and magnetic hyperthermia applications. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:108. [PMID: 34432156 PMCID: PMC8387263 DOI: 10.1007/s10856-021-06563-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/07/2021] [Indexed: 05/31/2023]
Abstract
Surface modification of superparamagnetic Fe3O4 nanoparticles using polymers (polyaniline/polypyrrole) was done by radio frequency (r.f.) plasma polymerization technique and characterized by XRD, TEM, TG/DTA and VSM. Surface-passivated Fe3O4 nanoparticles with polymers were having spherical/rod-shaped structures with superparamagnetic properties. Broad visible photoluminescence emission bands were observed at 445 and 580 nm for polyaniline-coated Fe3O4 and at 488 nm for polypyrrole-coated Fe3O4. These samples exhibit good fluorescence emissions with L929 cellular assay and were non-toxic. Magnetic hyperthermia response of Fe3O4 and polymer (polyaniline/polypyrrole)-coated Fe3O4 was evaluated and all the samples exhibit hyperthermia activity in the range of 42-45 °C. Specific loss power (SLP) values of polyaniline and polypyrrole-coated Fe3O4 nanoparticles (5 and 10 mg/ml) exhibit a controlled heat generation with an increase in the magnetic field.
Collapse
Affiliation(s)
- Beena Mol
- Department of Physics, Cochin University of Science and Technology, Cochin, Kerala, 682022, India
| | - Ansar Ereath Beeran
- Bioceramics Laboratory, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Poojappura, Kerala, 695012, India
- Department of Chemistry, M.E.S Asmabi College, P. Vemballur, Kodungallur, Thrissur, Kerala, 680671, India
| | - Prasad S Jayaram
- Division of Biophotonics and Imaging, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Poojappura, Thiruvananthapuram, Kerala, 695012, India
| | - Prabha Prakash
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin, Kerala, 682022, India
- Department of Biotechnology, Cochin University of Science and Technology, Cochin, Kerala, 682022, India
| | - Ramapurath S Jayasree
- Division of Biophotonics and Imaging, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Poojappura, Thiruvananthapuram, Kerala, 695012, India
| | - Senoy Thomas
- Department of Physics, Cochin University of Science and Technology, Cochin, Kerala, 682022, India
| | - Baby Chakrapani
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin, Kerala, 682022, India
- Department of Biotechnology, Cochin University of Science and Technology, Cochin, Kerala, 682022, India
| | - M R Anantharaman
- Department of Physics, Cochin University of Science and Technology, Cochin, Kerala, 682022, India.
| | - M Junaid Bushiri
- Department of Physics, Cochin University of Science and Technology, Cochin, Kerala, 682022, India.
| |
Collapse
|
31
|
Weeber R, Kreissl P, Holm C. Magnetic field controlled behavior of magnetic gels studied using particle-based simulations. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2019-0106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Abstract
This contribution provides an overview of the study of soft magnetic materials using particle-based simulation models. We focus in particular on systems where thermal fluctuations are important. As a basis for further discussion, we first describe two-dimensional models which demonstrate two deformation mechanisms of magnetic gels in a homogeneous field. One is based on the change of magnetic interactions between magnetic particles as a response to an external field; the other is the result of magnetically blocked particles acting as cross-linkers. Based on the qualitative behavior directly observable in the two-dimensional models, we extend our description to three-dimensions. We begin with particle-cross-linked gels, as for those, our three-dimensional model also includes explicitly resolved polymer chains. Here, the polymer chains are represented by entropic springs, and the deformation of the gel is the result of the interaction between magnetic particles. We use this model to examine the influence of the magnetic spatial configuration of magnetic particles (uniaxial or isotropic) on the gel’s magnetomechanical behavior. A further part of the article will be dedicated to scale-bridging approaches such as systematic coarse-graining and models located at the boundary between particle-based and continuum modeling. We will conclude our article with a discussion of recent results for modeling time-dependent phenomena in magnetic-polymer composites. The discussion will be focused on a simulation model suitable for obtaining AC-susceptibility spectra for dilute ferrofluids including hydrodynamic interactions. This model will be the basis for studying the signature of particle–polymer coupling in magnetic hybrid materials. In the long run, we aim to compare material properties probed locally via the AC-susceptibility spectra to elastic moduli obtained for the system at a global level.
Collapse
Affiliation(s)
- Rudolf Weeber
- Institute for Computational Physics, University of Stuttgart , Stuttgart , Germany
| | - Patrick Kreissl
- Institute for Computational Physics, University of Stuttgart , Stuttgart , Germany
| | - Christian Holm
- Institute for Computational Physics, University of Stuttgart , Stuttgart , Germany
| |
Collapse
|
32
|
Unak P, Hepton R, Harper M, Yasakci V, Pearce G, Russell S, Aras O, Akin O, Wong J. Toxicity testing of indocyanine green and fluorodeoxyglucose conjugated iron oxide nanoparticles with and without exposure to a magnetic field. ASIAN JOURNAL OF NANOSCIENCE AND MATERIALS 2021; 4:229-239. [PMID: 38192303 PMCID: PMC10773553 DOI: 10.26655/ajnanomat.2021.3.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Iron nanoparticles (MNPs) are known to induce membrane damage and apoptosis of cancer cells. In our study we determined whether FDG coupled with iron oxide magnetic nanoparticles can exert the same destructive effect on cancer cells. This research study presents data involving NIC-H727 human lung, bronchus epithelial cells exposed to conjugated fluorodeoxyglucose conjugated with iron-oxide magnetic nanoparticles and indocyanine green (ICG) dye (FDG-MNP-ICG), with and without the application of a magnetic field. Cell viability inferred from MTT assay revealed that FDG-MNPs had no significant toxicity towards noncancerous NIC-H727 human lung, bronchus epithelial cells. However, percentage cell death was much higher using a magnetic field, for the concentration of FDG-MNP-ICC used in our experiments. Magnetic field was able to destroy cells containing MNPs, while MNPs alone had significantly lower effects. Additionally, MNPs alone in these low concentrations had less adverse effects on healthy (non-target) cells.
Collapse
Affiliation(s)
- Perihan Unak
- Ege University, Institute of Nuclear Sciences, Department of Nuclear Applications, Bornova Izmir, 35100, Turkey
| | - Rachel Hepton
- Aston University, School of Life and Health Sciences, Aston Triangle, Birmingham, B4 7ET, United Kingdom
| | - Max Harper
- Aston University, School of Engineering and Applied Sciences, Aston Triangle, Birmingham, B4 7ET, United Kingdom
| | - Volkan Yasakci
- Ege University, Institute of Nuclear Sciences, Department of Nuclear Applications, Bornova Izmir, 35100, Turkey
| | - Gillian Pearce
- Aston University, School of Engineering and Applied Sciences, Aston Triangle, Birmingham, B4 7ET, United Kingdom
| | - Steve Russell
- Aston University, School of Life and Health Sciences, Aston Triangle, Birmingham, B4 7ET, United Kingdom
| | - Omer Aras
- Memorial Sloan Kettering Cancer Centre, Department of Radiology, New York, USA
| | - Oguz Akin
- Memorial Sloan Kettering Cancer Centre, Department of Radiology, New York, USA
| | - Julian Wong
- University Hospital Singapore, Cardiothoracic and Vascular Surgery Department, Singapore
| |
Collapse
|
33
|
Kundu P, Singh D, Singh A, Sahoo SK. Cancer Nanotheranostics: A Nanomedicinal Approach for Cancer Therapy and Diagnosis. Anticancer Agents Med Chem 2021; 20:1288-1299. [PMID: 31429694 DOI: 10.2174/1871520619666190820145930] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 12/27/2022]
Abstract
The panorama of cancer treatment has taken a considerable leap over the last decade with the advancement in the upcoming novel therapies combined with modern diagnostics. Nanotheranostics is an emerging science that holds tremendous potential as a contrivance by integrating therapy and imaging in a single probe for cancer diagnosis and treatment thus offering the advantage like tumor-specific drug delivery and at the same time reduced side effects to normal tissues. The recent surge in nanomedicine research has also paved the way for multimodal theranostic nanoprobe towards personalized therapy through interaction with a specific biological system. This review presents an overview of the nano theranostics approach in cancer management and a series of different nanomaterials used in theranostics and the possible challenges with future directions.
Collapse
Affiliation(s)
- Paromita Kundu
- Laboratory of Nanomedicine, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - Deepika Singh
- Laboratory of Nanomedicine, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - Abhalaxmi Singh
- Laboratory of Nanomedicine, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - Sanjeeb K Sahoo
- Laboratory of Nanomedicine, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| |
Collapse
|
34
|
Khan MA, Singh D, Ahmad A, Siddique HR. Revisiting inorganic nanoparticles as promising therapeutic agents: A paradigm shift in oncological theranostics. Eur J Pharm Sci 2021; 164:105892. [PMID: 34052295 DOI: 10.1016/j.ejps.2021.105892] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/16/2022]
Abstract
Cancer remains a global health problem largely due to a lack of effective therapies. Major cancer management strategies include chemotherapy, surgical resection, and radiation. Unfortunately, these strategies have a number of limitations, such as non-specific side effects, uneven delivery of the drugs, and lack of proper monitoring technology. Inorganic nanoparticles (NPs) are considered promising agents in treating and tracing cancer due to their unique physicochemical properties such as the controlled release of drugs, bioavailability, biocompatibility, stability, and large surface area. Also, they enhance the solubility of hydrophobic drugs, prolong their circulation time, prevent undesired off-targeting and subsequent side effects, making them efficient particles in cancer theranostics. Promising inorganic-NPs include gold, selenium, silica, and oxide NPs. Further, several techniques are used to modify the surface of inorganic-NPs, making them more efficient for the effective transport of therapeutic cargos to overcome cellular barriers. Thus, inorganic-NPs function effectively, surmounting the intrinsic drawbacks of traditional organic NPs. This mini-review summarizes the significant inorganic-NPs, their properties, surface modifications, cellular uptake, and bio-distributions, along with their potential use in cancer theranostics. We also discuss the promises and challenges faced during the inorganic-NPs mediated therapeutic approach for cancer and these particles' status in the clinical setting.
Collapse
Affiliation(s)
- Mohammad Afsar Khan
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India
| | - Deepti Singh
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India
| | - Absar Ahmad
- Interdisciplinary Nanotechnology Centre, Aligarh Muslim University, Aligarh, 202002, India
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India
| |
Collapse
|
35
|
Xuan Y, Guan M, Zhang S. Tumor immunotherapy and multi-mode therapies mediated by medical imaging of nanoprobes. Theranostics 2021; 11:7360-7378. [PMID: 34158855 PMCID: PMC8210602 DOI: 10.7150/thno.58413] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/14/2021] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy is an effective tumor treatment strategy that has several advantages over conventional methods such as surgery, radiotherapy and chemotherapy. Studies show that multifunctional nanoprobes can achieve multi-mode image-guided multiple tumor treatment modes. The tumor cells killed by chemotherapies or phototherapies release antigens that trigger an immune response and augment the effects of tumor immunotherapy. Thus, combining immunotherapy and multifunctional nanoprobes can achieve early cancer diagnosis and treatment. In this review, we have summarized the current research on the applications of multifunctional nanoprobes in image-guided immunotherapy. In addition, image-guided synergistic chemotherapy/photothermal therapy/photodynamic therapy and immunotherapy have also been discussed. Furthermore, the application potential and clinical prospects of multifunctional nanoprobes in combination with immunotherapy have been assessed.
Collapse
Affiliation(s)
| | | | - Shubiao Zhang
- Key Lab of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, 116600, China
| |
Collapse
|
36
|
Advances in nano-biomaterials and their applications in biomedicine. Emerg Top Life Sci 2021; 5:169-176. [PMID: 33825835 DOI: 10.1042/etls20200333] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 01/26/2023]
Abstract
Nanotechnology has received considerable attention and interest over the past few decades in the field of biomedicine due to the wide range of applications it provides in disease diagnosis, drug design and delivery, biomolecules detection, tissue engineering and regenerative medicine. Ultra-small size and large surface area of nanomaterials prove to be greatly advantageous for their biomedical applications. Moreover, the physico-chemical and thus, the biological properties of nanomaterials can be manipulated depending on the application. However, stability, efficacy and toxicity of nanoparticles remain challenge for researchers working in this area. This mini-review highlights the recent advances of various types of nanoparticles in biomedicine and will be of great value to researchers in the field of materials science, chemistry, biology and medicine.
Collapse
|
37
|
Ou YH, Liang J, Czarny B, Wacker MG, Yu V, Wang JW, Pastorin G. Extracellular Vesicle (EV) biohybrid systems for cancer therapy: Recent advances and future perspectives. Semin Cancer Biol 2021; 74:45-61. [PMID: 33609664 DOI: 10.1016/j.semcancer.2021.02.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/10/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are a class of cell-derived lipid-bilayer membrane vesicles secreted by almost all mammalian cells and involved in intercellular communication by shuttling various biological cargoes. Over the last decade, EVs - namely exosomes and microvesicles - have been extensively explored as next-generation nanoscale drug delivery systems (DDSs). This is in large due to their endogenous origin, which enables EVs to circumvent some of the limitations associated with existing cancer therapy approaches (i.e. by preventing recognition by the immune system and improving selectivity towards tumor tissue). However, successful translation of these cell-derived vesicles into clinical applications has been hindered by several factors, among which the loading of exogenous therapeutic molecules still represents a great challenge. In order to address this issue and to further advance these biologically-derived systems as drug carriers, EV-biohybrid nano-DDSs, obtained through the fusion of EVs with conventional synthetic nano-DDSs, have recently been proposed as a valuable alternative as DDSs. Building on the idea of "combining the best of both worlds", a combination of these two unique entities aims to harness the beneficial properties associated with both EVs and conventional nano-DDSs, while overcoming the flaws of the individual components. These biohybrid systems also provide a unique opportunity for exploitation of new synergisms, often leading to improved therapeutic outcomes, thus paving the way for advancements in cancer therapy. This review aims to describe the recent developments of EV-biohybrid nano-DDSs in cancer therapy, to highlight the most promising results and breakthroughs, as well as to provide a glimpse on the possible intrinsic targeting mechanisms of EVs that can be bequeathed to their hybrid systems. Finally, we also provide some insights in the future perspectives of EV-hybrid DDSs.
Collapse
Affiliation(s)
- Yi-Hsuan Ou
- Department of Pharmacy, National University of Singapore, Singapore
| | - Jeremy Liang
- Department of Pharmacy, National University of Singapore, Singapore
| | - Bertrand Czarny
- School of Materials Science & Engineering and Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | | | - Victor Yu
- Department of Pharmacy, National University of Singapore, Singapore
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Cardiovascular Research Institute, National University Heart Centre, Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Giorgia Pastorin
- Department of Pharmacy, National University of Singapore, Singapore.
| |
Collapse
|
38
|
Taufiq A, Saputro RE, Susanto H, Hidayat N, Sunaryono S, Amrillah T, Wijaya HW, Mufti N, Simanjuntak FM. Synthesis of Fe 3O 4/Ag nanohybrid ferrofluids and their applications as antimicrobial and antifibrotic agents. Heliyon 2020; 6:e05813. [PMID: 33426329 PMCID: PMC7779699 DOI: 10.1016/j.heliyon.2020.e05813] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/25/2020] [Accepted: 12/18/2020] [Indexed: 12/23/2022] Open
Abstract
To date, the search for creating stable ferrofluids with excellent properties for biomedical application is one of the challenging scientific and practical investigations. In this study, novel Fe3O4/Ag nanohybrid ferrofluids from iron sand were synthesized using a double-layer method. The Fe3O4/Ag nanocomposites exhibited stable crystallite sizes of 11.8 12.1 nm and 36.8-37.2 nm for Fe3O4 and Ag, respectively. The lattice parameters of the spinel structure Fe3O4 and face-centered cubic Ag were respectively 8.344 Å and 4.091 Å. With increasing Ag amount, the crystallite phase of Ag in the nanocomposites increased from 40.2% to 77.2%. The XPS results confirmed that Fe3O4/Ag nanocomposites were successfully prepared, where Fe3O4 mixed well with Ag via strong ionic bonding. The FTIR results confirmed the presence of Fe3O4/Ag, oleic acid, and dimethyl sulfoxide as the filler, first layer, and second layer, respectively. The as-prepared ferrofluids exhibited superparamagnetic behavior, where the saturation magnetization decreased with increasing Ag content. The Fe3O4/Ag nanohybrid ferrofluids exhibited excellent antimicrobial performance against Bacillus subtilis, Staphylococcus aureus, Escherichia coli, and Candida albicans. More importantly, the Fe3O4/Ag nanohybrid ferrofluids decreased the progression of liver fibrosis-related inflammation and fibrogenic activity on hepatic stellate cells.
Collapse
Affiliation(s)
- Ahmad Taufiq
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Negeri Malang, Jl. Semarang 5, Malang, 65145, Indonesia
| | - Rosy Eko Saputro
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Negeri Malang, Jl. Semarang 5, Malang, 65145, Indonesia
| | - Hendra Susanto
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Negeri Malang, Jl. Semarang 5, Malang, 65145, Indonesia
| | - Nurul Hidayat
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Negeri Malang, Jl. Semarang 5, Malang, 65145, Indonesia
| | - Sunaryono Sunaryono
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Negeri Malang, Jl. Semarang 5, Malang, 65145, Indonesia
| | - Tahta Amrillah
- Department of Physics, Faculty of Science and Technology, Universitas Airlangga, Surabaya, 60115, Indonesia
| | - Husni Wahyu Wijaya
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Negeri Malang, Jl. Semarang 5, Malang, 65145, Indonesia
| | - Nandang Mufti
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Negeri Malang, Jl. Semarang 5, Malang, 65145, Indonesia
| | - Firman Mangasa Simanjuntak
- Zepler Institute for Photonics and Nanoelectronics, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| |
Collapse
|
39
|
Albukhaty S, Al-Musawi S, Abdul Mahdi S, Sulaiman GM, Alwahibi MS, Dewir YH, Soliman DA, Rizwana H. Investigation of Dextran-Coated Superparamagnetic Nanoparticles for Targeted Vinblastine Controlled Release, Delivery, Apoptosis Induction, and Gene Expression in Pancreatic Cancer Cells. Molecules 2020; 25:molecules25204721. [PMID: 33076247 PMCID: PMC7587551 DOI: 10.3390/molecules25204721] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
In the current study, the surface of superparamagnetic iron oxide (SPION) was coated with dextran (DEX), and conjugated with folic acid (FA), to enhance the targeted delivery and uptake of vinblastine (VBL) in PANC-1 pancreatic cancer cells. Numerous analyses were performed to validate the prepared FA-DEX-VBL-SPION, such as field emission scanning transmission electron microscopy, high-resolution transmission electron microscopy, dynamic light scattering (DLS), Zeta Potential, Fourier transform infrared spectroscopy, and vibrating sample magnetometry (VSM). The delivery system capacity was evaluated by loading and release experiments. Moreover, in vitro biological studies, including a cytotoxicity study, cellular uptake assessment, apoptosis analysis, and real-time PCR, were carried out. The results revealed that the obtained nanocarrier was spherical with a suitable dispersion and without visible aggregation. Its average size, polydispersity, and zeta were 74 ± 13 nm, 0.080, and −45 mV, respectively. This dual functional nanocarrier also exhibited low cytotoxicity and a high apoptosis induction potential for successful VBL co-delivery. Real-time quantitative PCR analysis demonstrated the activation of caspase-3, NF-1, PDL-1, and H-ras inhibition, in PANC-1 cells treated with the FA-VBL-DEX-SPION nanostructure. Close inspection of the obtained data proved that the FA-VBL-DEX-SPION nanostructure possesses a noteworthy chemo-preventive effect on pancreatic cancer cells through the inhibition of cell proliferation and induction of apoptosis.
Collapse
Affiliation(s)
- Salim Albukhaty
- Department of Basic Sciences, College of Nursing, University of Misan, Maysan 62001, Iraq;
| | - Sharafaldin Al-Musawi
- Faculty of Biotechnology, Al-Qasim Green University, Babylon 51013, Iraq;
- Correspondence:
| | - Salih Abdul Mahdi
- Faculty of Biotechnology, Al-Qasim Green University, Babylon 51013, Iraq;
| | - Ghassan M. Sulaiman
- Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq;
| | - Mona S. Alwahibi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.S.A.); (D.A.S.); (H.R.)
| | - Yaser Hassan Dewir
- College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia;
- Faculty of Agriculture, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Dina A. Soliman
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.S.A.); (D.A.S.); (H.R.)
| | - Humaira Rizwana
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.S.A.); (D.A.S.); (H.R.)
| |
Collapse
|
40
|
Liu TI, Lu TY, Yang YC, Chang SH, Chen HH, Lu IL, Sabu A, Chiu HC. New combination treatment from ROS-Induced sensitized radiotherapy with nanophototherapeutics to fully eradicate orthotopic breast cancer and inhibit metastasis. Biomaterials 2020; 257:120229. [DOI: 10.1016/j.biomaterials.2020.120229] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
|
41
|
Mitoxantrone-Loaded Nanoparticles for Magnetically Controlled Tumor Therapy-Induction of Tumor Cell Death, Release of Danger Signals and Activation of Immune Cells. Pharmaceutics 2020; 12:pharmaceutics12100923. [PMID: 32992645 PMCID: PMC7599695 DOI: 10.3390/pharmaceutics12100923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/17/2020] [Accepted: 09/23/2020] [Indexed: 12/30/2022] Open
Abstract
Stimulating the patient’s immune system represents a promising therapeutic strategy to fight cancer. However, low immunogenicity of the tumor cells within an immune suppressive milieu often leads to weak anti-tumor immune responses. Additionally, the immune system may be impaired by accompanying aggressive chemotherapies. We show that mitoxantrone, bound to superparamagnetic iron oxide nanoparticles (SPIONs) as the transport system, can be magnetically accumulated in adherent HT-29 colon carcinoma cells, thereby inducing the same cell death phenotype as its soluble counterpart, a chemotherapeutic agent and prototypic inductor of immunogenic cell death. The nanoparticle-loaded drug induces cell cycle stop, apoptosis and secondary necrosis in a dose- and time-dependent manner comparable to the free drug. Cell death was accompanied by the release of interleukin-8 and damage-associated molecular patterns (DAMPs) such as HSP70 and ATP, which fostered chemotactic migration of monocytes and maturation of dendritic cells. We furthermore ensured absence of endotoxin contaminations and compatibility with erythrocytes and platelets and investigated the influence on plasma coagulation in vitro. Summarizing, with magnetic enrichment, mitoxantrone can be accumulated at the desired place, sparing healthy peripheral cells and tissues, such as immune cells. Conserving immune competence in cancer patients in the future might allow combined therapeutic approaches with immune therapies (e.g., checkpoint inhibitors).
Collapse
|
42
|
Patil-Sen Y, Torino E, De Sarno F, Ponsiglione AM, Chhabria V, Ahmed W, Mercer T. Biocompatible superparamagnetic core-shell nanoparticles for potential use in hyperthermia-enabled drug release and as an enhanced contrast agent. NANOTECHNOLOGY 2020; 31:375102. [PMID: 32392545 DOI: 10.1088/1361-6528/ab91f6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) and core-shell type nanoparticles, consisting of SPIONs coated with mesoporous silica and/or lipid, were synthesised and tested for their potential theranostic applications in drug delivery, magnetic hyperthermia and as a contrast agent. Transmission Electron Microscopy (TEM) confirmed the size of bare and coated SPIONs was in the range of 5-20 nm and 100-200 nm respectively. The superparamagnetic nature of all the prepared nanomaterials as indicated by Vibrating Sample Magnetometry (VSM) and their heating properties under an AC field confirm their potential for hyperthermia applications. Scanning Column Magnetometry (SCM) data showed that extrusion of bare-SPION (b-SPION) dispersions through a 100 nm polycarbonate membrane significantly improved the dispersion stability of the sample. No sedimentation was apparent after 18 h compared to a pre-extrusion estimate of 43% settled at the bottom of the tube over the same time. Lipid coating also enhanced dispersion stability. Transversal relaxation time (T2) measurements for the nanoparticles, using a bench-top relaxometer, displayed a significantly lower value of 46 ms, with a narrow relaxation time distribution, for lipid silica coated SPIONs (Lip-SiSPIONs) as compared to that of 1316 ms for the b-SPIONs. Entrapment efficiency of the anticancer drug, Doxorubicin (DOX) for Lip-SPIONs was observed to be 35% which increased to 58% for Lip-SiSPIONs. Moreover, initial in-vitro cytotoxicity studies against human breast adenocarcinoma, MCF-7 cells showed that % cell viability increased from 57% for bSPIONs to 82% for Lip-SPIONs and to 87% for Lip-SiSPIONs. This suggests that silica and lipid coatings improve the biocompatibility of bSPIONs significantly and enhance the suitability of these particles as drug carriers. Hence, the magnetic nanomaterials prepared in this work have potential theranostic properties as a drug carrier for hyperthermia cancer therapy and also offer enhancement of contrast agent efficacy and a route to a significant increase in dispersion stability.
Collapse
Affiliation(s)
- Yogita Patil-Sen
- School of Physical Sciences and Computing, University of Central Lancashire, Preston PR1 2HE, United Kingdom. School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
43
|
Rueda-Gensini L, Cifuentes J, Castellanos MC, Puentes PR, Serna JA, Muñoz-Camargo C, Cruz JC. Tailoring Iron Oxide Nanoparticles for Efficient Cellular Internalization and Endosomal Escape. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1816. [PMID: 32932957 PMCID: PMC7559083 DOI: 10.3390/nano10091816] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022]
Abstract
Iron oxide nanoparticles (IONs) have been widely explored for biomedical applications due to their high biocompatibility, surface-coating versatility, and superparamagnetic properties. Upon exposure to an external magnetic field, IONs can be precisely directed to a region of interest and serve as exceptional delivery vehicles and cellular markers. However, the design of nanocarriers that achieve an efficient endocytic uptake, escape lysosomal degradation, and perform precise intracellular functions is still a challenge for their application in translational medicine. This review highlights several aspects that mediate the activation of the endosomal pathways, as well as the different properties that govern endosomal escape and nuclear transfection of magnetic IONs. In particular, we review a variety of ION surface modification alternatives that have emerged for facilitating their endocytic uptake and their timely escape from endosomes, with special emphasis on how these can be manipulated for the rational design of cell-penetrating vehicles. Moreover, additional modifications for enhancing nuclear transfection are also included in the design of therapeutic vehicles that must overcome this barrier. Understanding these mechanisms opens new perspectives in the strategic development of vehicles for cell tracking, cell imaging and the targeted intracellular delivery of drugs and gene therapy sequences and vectors.
Collapse
Affiliation(s)
- Laura Rueda-Gensini
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Javier Cifuentes
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Maria Claudia Castellanos
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Paola Ruiz Puentes
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Julian A. Serna
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide 5005, Australia
| |
Collapse
|
44
|
Klapproth AP, Shevtsov M, Stangl S, Li WB, Multhoff G. A New Pharmacokinetic Model Describing the Biodistribution of Intravenously and Intratumorally Administered Superparamagnetic Iron Oxide Nanoparticles (SPIONs) in a GL261 Xenograft Glioblastoma Model. Int J Nanomedicine 2020; 15:4677-4689. [PMID: 32669844 PMCID: PMC7335747 DOI: 10.2147/ijn.s254745] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/21/2020] [Indexed: 12/22/2022] Open
Abstract
Background Superparamagnetic iron oxide nanoparticles (SPIONs) have displayed multifunctional applications in cancer theranostics following systemic delivery. In an effort to increase the therapeutic potential of local therapies (including focal hyperthermia), nanoparticles can also be administered intratumorally. Therefore, the development of a reliable pharmacokinetic model for the prediction of nanoparticle distribution for both clinically relevant routes of delivery is of high importance. Materials and Methods The biodistribution of SPIONs (of two different sizes – 130 nm and 60 nm) radiolabeled with zirconium-89 or technetium-99m following intratumoral or intravenous injection was investigated in C57/Bl6 mice bearing subcutaneous GL261 glioblastomas. Based on PET/CT biodistribution data, a novel pharmacokinetic model was established for a better understanding of the pharmacokinetics of the SPIONs after both administration routes. Results The PET image analysis of the nanoparticles (confirmed by histology) demonstrated the presence of radiolabeled nanoparticles within the glioma site (with low amounts in the liver and spleen) at all investigated time points following intratumoral injection. The mathematical model confirmed the dynamic nanoparticle redistribution in the organism over a period of 72 h with an equilibrium reached after 100 h. Intravenous injection of nanoparticles demonstrated a different distribution pattern with a rapid particle retention in all organs (particularly in liver and spleen) and a subsequent slow release rate. Conclusion The mathematical model demonstrated good agreement with experimental data derived from tumor mouse models suggesting the value of this tool to predict the real-time pharmacokinetic features of SPIONs in vivo. In the future, it is planned to adapt our model to other nanoparticle formulations to more precisely describe their biodistribution in in vivo model systems.
Collapse
Affiliation(s)
- Alexander P Klapproth
- Center for Translational Cancer Research Technische Universität München (TranslaTUM), Klinikum Rechts Der Isar, Munich, Germany.,Institute of Radiation Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Maxim Shevtsov
- Center for Translational Cancer Research Technische Universität München (TranslaTUM), Klinikum Rechts Der Isar, Munich, Germany.,Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia.,Department of Biotechnology, First Pavlov State Medical University of St. Petersburg, St. Petersburg, Russia.,Almazov National Medical Research Centre, Russian Polenov Neurosurgical Institute, St. Petersburg, Russia.,National Center for Neurosurgery, Nur-Sultan, Kazakhstan.,Department of Biomedical Cell Technologies, Far Eastern Federal University, Vladivostok, Russia
| | - Stefan Stangl
- Center for Translational Cancer Research Technische Universität München (TranslaTUM), Klinikum Rechts Der Isar, Munich, Germany
| | - Wei Bo Li
- Institute of Radiation Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Gabriele Multhoff
- Center for Translational Cancer Research Technische Universität München (TranslaTUM), Klinikum Rechts Der Isar, Munich, Germany
| |
Collapse
|
45
|
Iron Oxide/Salicylic Acid Nanoparticles as Potential Therapy for B16F10 Melanoma Transplanted on the Chick Chorioallantoic Membrane. Processes (Basel) 2020. [DOI: 10.3390/pr8060706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Unfavorable prognoses and low survival rates are specific features of metastatic melanoma that justify the concern for the development of new therapeutic strategies. Lately, nanotechnology has become an attractive field of study due to recent advances in nanomedicine. Using a chick chorioallantoic membrane (CAM) implanted with xenografts harvested from C57BL/6 mice with B16F10 melanoma cells, we studied the effects of iron oxide nanoparticles functionalized with salicylic acid (SaMNPs) as a form of therapy on the local development of xenotransplants and CAM vessels. The SaMNPs induced an anti-angiogenic effect on the CAM vessels, which accumulated preferentially in the melanoma cells and induced apoptosis and extensive xenograft necrosis. As a result, this slowed the increase in the xenograft volume and reduced the melanoma cells’ ability to metastasize locally and distally. Further, we demonstrate the use of the chick CAM model as a tool for testing the action of newly synthesized nanocomposites on melanoma xenotransplants. The SaMNPs had a therapeutic effect on B16F10 melanoma due to the synergistic action of the two components of its structure: the coating of the salicylic acid with antiangiogenic and chemotherapeutic action and the core of iron oxides with cytotoxic action.
Collapse
|
46
|
Putz AM, Ianăși C, Dudás Z, Coricovac D, Watz C(F, Len A, Almásy L, Sacarescu L, Dehelean C. SiO 2-PVA-Fe(acac) 3 Hybrid Based Superparamagnetic Nanocomposites for Nanomedicine: Morpho-textural Evaluation and In Vitro Cytotoxicity Assay. Molecules 2020; 25:molecules25030653. [PMID: 32033018 PMCID: PMC7038086 DOI: 10.3390/molecules25030653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/31/2020] [Accepted: 02/01/2020] [Indexed: 12/29/2022] Open
Abstract
A facile sol-gel route has been applied to synthesize hybrid silica-PVA-iron oxide nanocomposite materials. A step-by-step calcination (processing temperatures up to 400 °C) was applied in order to oxidize the organics together with the iron precursor. Transmission electron microscopy, X-ray diffraction, small angle neutron scattering, and nitrogen porosimetry were used to determine the temperature-induced morpho-textural modifications. In vitro cytotoxicity assay was conducted by monitoring the cell viability by the means of MTT assay to qualify the materials as MRI contrast agents or as drug carriers. Two cell lines were considered: the HaCaT (human keratinocyte cell line) and the A375 tumour cell line of human melanoma. Five concentrations of 10 µg/mL, 30 µg/mL, 50 µg/mL, 100 µg/mL, and 200 µg/mL were tested, while using DMSO (dimethylsulfoxid) and PBS (phosphate saline buffer) as solvents. The HaCaT and A375 cell lines were exposed to the prepared agent suspensions for 24 h. In the case of DMSO (dimethyl sulfoxide) suspensions, the effect on human keratinocytes migration and proliferation were also evaluated. The results indicate that only the concentrations of 100 μg/mL and 200 μg/mL of the nanocomposite in DMSO induced a slight decrease in the HaCaT cell viability. The PBS based in vitro assay showed that the nanocomposite did not present toxicity on the HaCaT cells, even at high doses (200 μg/mL agent).
Collapse
Affiliation(s)
- Ana-Maria Putz
- ”Coriolan Dragulescu” Institute of Chemistry, Romanian Academy, Mihai Viteazul Bd., No. 24, 300223 Timişoara, Romania; (A.-M.P.); (C.I.)
| | - Cătălin Ianăși
- ”Coriolan Dragulescu” Institute of Chemistry, Romanian Academy, Mihai Viteazul Bd., No. 24, 300223 Timişoara, Romania; (A.-M.P.); (C.I.)
| | - Zoltán Dudás
- Wigner Research Centre for Physics, POB 49 1525 Budapest, Hungary
- Correspondence:
| | - Dorina Coricovac
- Pharmacy II Department, Faculty of Pharmacy, “Victor Babes ¸” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (D.C.)
| | - Claudia (Farcas) Watz
- Pharmacy II Department, Faculty of Pharmacy, “Victor Babes ¸” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (D.C.)
| | - Adél Len
- Centre for Energy Research, Konkoly-Thege 29-33, 1121 Budapest, Hungary;
- University of Pécs, Faculty of Engineering and Information technology, Boszorkány St. 2, 7624 Pécs, Hungary
| | - László Almásy
- Wigner Research Centre for Physics, POB 49 1525 Budapest, Hungary
| | - Liviu Sacarescu
- Institute of Macromolecular Chemistry “Petru Poni”, Aleea Grigore Ghica Voda, nr. 41A 700487 Iasi, Romania;
| | - Cristina Dehelean
- Pharmacy II Department, Faculty of Pharmacy, “Victor Babes ¸” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (D.C.)
| |
Collapse
|
47
|
Ferraz FS, López JL, Lacerda SMSN, Procópio MS, Figueiredo AFA, Martins EMN, Guimarães PPG, Ladeira LO, Kitten GT, Dias FF, Domingues RZ, Costa GMJ. Biotechnological approach to induce human fibroblast apoptosis using superparamagnetic iron oxide nanoparticles. J Inorg Biochem 2020; 206:111017. [PMID: 32120160 DOI: 10.1016/j.jinorgbio.2020.111017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 01/06/2023]
Abstract
Cancer-Associated Fibroblasts (CAFs) contribute to tumour progression and have received significant attention as a therapeutic target. These cells produce growth factors, cytokines and chemokines, stimulating cancer cell proliferation and inhibiting their apoptosis. Recent advances in drug delivery have demonstrated a significant promise of iron oxide nanoparticles in clinics as theranostic agents, mainly due to their magnetic properties. Here, we designed superparamagnetic iron oxide nanoparticles (SPIONs) to induce apoptosis of human fibroblasts. SPIONs were synthesized via co-precipitation method and coated with sodium citrate (SPION_Cit). We assessed the intracellular uptake of SPIONs by human fibroblast cells, as well as their cytotoxicity and ability to induce thermal effects under the magnetic field. The efficiency and time of nanoparticle internalization were assessed by Prussian Blue staining, flow cytometry and transmission electron microscopy. SPIONs_Cit were detected in the cytoplasm of human fibroblasts 15 min after in vitro exposure, entering into cells mainly via endocytosis. Analyses through Cell Titer Blue assay, AnnexinV-fluorescein isothiocyanate (FITC) and propidium iodide (PI) cellular staining demonstrated that concentrations below 8 × 10-2 mg/mL of SPIONs_Cit did not alter cell viability of human fibroblast. Furthermore, it was also demonstrated that SPIONs_Cit associated with alternating current magnetic field were able to induce hyperthermia and human fibroblast cell death in vitro, mainly through apoptosis (83.5%), activating caspase 8 (extrinsic apoptotic via) after a short exposure period. Collectively these findings suggest that our nanoplatform is biocompatible and can be used for therapeutic purposes in human biological systems, such as inducing apoptosis of CAFs.
Collapse
Affiliation(s)
- Fausto S Ferraz
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jorge L López
- Center for Biological and Natural Sciences, Federal University of Acre, Rio Branco, AC, Brazil
| | - Samyra M S N Lacerda
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcela S Procópio
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - André F A Figueiredo
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Estefânia M N Martins
- Laboratory of Chemistry of Nanostructures, Nuclear Technology Development Center, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro P G Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiz O Ladeira
- Laboratory of Nanomaterials, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gregory T Kitten
- Microscopy Center, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Felipe F Dias
- Microscopy Center, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rosana Z Domingues
- Laboratory of Chemistry, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Guilherme M J Costa
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
48
|
Turin-Moleavin IA, Fifere A, Lungoci AL, Rosca I, Coroaba A, Peptanariu D, Nastasa V, Pasca SA, Bostanaru AC, Mares M, Pinteala M. In Vitro and In Vivo Antioxidant Activity of the New Magnetic-Cerium Oxide Nanoconjugates. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1565. [PMID: 31690040 PMCID: PMC6915648 DOI: 10.3390/nano9111565] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 10/21/2019] [Accepted: 11/01/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND Cerium oxide nanoparticles present the mimetic activity of superoxide dismutase, being able to inactivate the excess of reactive oxygen species (ROS) correlated with a large number of pathologies, such as stents restenosis and the occurrence of genetic mutations that can cause cancer. This study presents the synthesis and biological characterisation of nanoconjugates based on nanoparticles of iron oxide interconnected with cerium oxide conjugates. METHODS The synthesis of magnetite-nanoceria nanoconjugates has been done in several stages, where the key to the process is the coating of nanoparticles with polyethyleneimine and its chemical activation-reticulation with glutaraldehyde. The nanoconjugates are characterised by several techniques, and the antioxidant activity was evaluated in vitro and in vivo. RESULTS Iron oxide nanoparticles interconnected with cerium oxide nanoparticles were obtained, having an average diameter of 8 nm. Nanoconjugates prove to possess superparamagnetic properties and the saturation magnetisation varies with the addition of diamagnetic components in the system, remaining within the limits of biomedical applications. In vitro free-radical scavenging properties of nanoceria are improved after the coating of nanoparticles with polyethylenimine and conjugation with magnetite nanoparticles. In vivo studies reveal increased antioxidant activity in all organs and fluids collected from mice, which demonstrates the ability of the nanoconjugates to reduce oxidative stress. CONCLUSION Nanoconjugates possess magnetic properties, being able to scavenge free radicals, reducing the oxidative stress. The combination of the two properties mentioned above makes them excellent candidates for theranostic applications.
Collapse
Affiliation(s)
- Ioana-Andreea Turin-Moleavin
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica-Voda Alley, 700487 Iasi, Romania; (I.-A.T.-M.); (A.-L.L.); (I.R.); (A.C.); (D.P.)
| | - Adrian Fifere
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica-Voda Alley, 700487 Iasi, Romania; (I.-A.T.-M.); (A.-L.L.); (I.R.); (A.C.); (D.P.)
| | - Ana-Lacramioara Lungoci
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica-Voda Alley, 700487 Iasi, Romania; (I.-A.T.-M.); (A.-L.L.); (I.R.); (A.C.); (D.P.)
| | - Irina Rosca
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica-Voda Alley, 700487 Iasi, Romania; (I.-A.T.-M.); (A.-L.L.); (I.R.); (A.C.); (D.P.)
| | - Adina Coroaba
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica-Voda Alley, 700487 Iasi, Romania; (I.-A.T.-M.); (A.-L.L.); (I.R.); (A.C.); (D.P.)
| | - Dragos Peptanariu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica-Voda Alley, 700487 Iasi, Romania; (I.-A.T.-M.); (A.-L.L.); (I.R.); (A.C.); (D.P.)
| | - Valentin Nastasa
- Laboratory of Antimicrobial Chemotherapy, “Ion Ionescu de la Brad” University of Agricultural Sciences and Veterinary Medicine, 8 Sadoveanu Alley, 700489 Iasi, Romania; (V.N.); (S.-A.P.); (A.-C.B.); (M.M.)
| | - Sorin-Aurelian Pasca
- Laboratory of Antimicrobial Chemotherapy, “Ion Ionescu de la Brad” University of Agricultural Sciences and Veterinary Medicine, 8 Sadoveanu Alley, 700489 Iasi, Romania; (V.N.); (S.-A.P.); (A.-C.B.); (M.M.)
| | - Andra-Cristina Bostanaru
- Laboratory of Antimicrobial Chemotherapy, “Ion Ionescu de la Brad” University of Agricultural Sciences and Veterinary Medicine, 8 Sadoveanu Alley, 700489 Iasi, Romania; (V.N.); (S.-A.P.); (A.-C.B.); (M.M.)
| | - Mihai Mares
- Laboratory of Antimicrobial Chemotherapy, “Ion Ionescu de la Brad” University of Agricultural Sciences and Veterinary Medicine, 8 Sadoveanu Alley, 700489 Iasi, Romania; (V.N.); (S.-A.P.); (A.-C.B.); (M.M.)
| | - Mariana Pinteala
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica-Voda Alley, 700487 Iasi, Romania; (I.-A.T.-M.); (A.-L.L.); (I.R.); (A.C.); (D.P.)
| |
Collapse
|
49
|
Mukherjee S, Sonanini D, Maurer A, Daldrup-Link HE. The yin and yang of imaging tumor associated macrophages with PET and MRI. Am J Cancer Res 2019; 9:7730-7748. [PMID: 31695797 PMCID: PMC6831464 DOI: 10.7150/thno.37306] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022] Open
Abstract
Tumor associated macrophages (TAM) are key players in the cancer microenvironment. Molecular imaging modalities such as MRI and PET can be used to track and monitor TAM dynamics in tumors non-invasively, based on specific uptake and quantification of MRI-detectable nanoparticles or PET-detectable radiotracers. Particular molecular signatures can be leveraged to target anti-inflammatory TAM, which support tumor growth, and pro-inflammatory TAM, which suppress tumor growth. In addition, TAM-directed imaging probes can be designed to include immune modulating properties, thereby leading to combined diagnostic and therapeutic (theranostic) effects. In this review, we will discuss the complementary role of TAM-directed radiotracers and iron oxide nanoparticles for monitoring cancer immunotherapies with PET and MRI technologies. In addition, we will outline how TAM-directed imaging and therapy is interdependent and can be connected towards improved clinical outcomes
Collapse
|
50
|
Ferjaoui Z, Jamal Al Dine E, Kulmukhamedova A, Bezdetnaya L, Soon Chang C, Schneider R, Mutelet F, Mertz D, Begin-Colin S, Quilès F, Gaffet E, Alem H. Doxorubicin-Loaded Thermoresponsive Superparamagnetic Nanocarriers for Controlled Drug Delivery and Magnetic Hyperthermia Applications. ACS APPLIED MATERIALS & INTERFACES 2019; 11:30610-30620. [PMID: 31359758 DOI: 10.1021/acsami.9b10444] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
This study reports on the development of thermoresponsive core/shell magnetic nanoparticles (MNPs) based on an iron oxide core and a thermoresponsive copolymer shell composed of 2-(2-methoxy)ethyl methacrylate (MEO2MA) and oligo(ethylene glycol)methacrylate (OEGMA) moieties. These smart nano-objects combine the magnetic properties of the core and the drug carrier properties of the polymeric shell. Loading the anticancer drug doxorubicin (DOX) in the thermoresponsive MNPs via supramolecular interactions provides advanced features to the delivery of DOX with spatial and temporal controls. The so coated iron oxide MNPs exhibit superparamagnetic behavior with a saturation magnetization of around 30 emu g-1. Drug release experiments confirmed that only a small amount of DOX was released at room temperature, while almost 100% drug release was achieved after 52 h at 42 °C with Fe3-δO4@P(MEO2MA60OEGMA40), which grafted polymer chains displaying a low critical solution temperature of 41 °C. Moreover, the MNPs exhibit magnetic hyperthermia properties as shown by specific absorption rate measurements. Finally, the cytotoxicity of the core/shell MNPs toward human ovary cancer SKOV-3 cells was tested. The results showed that the polymer-capped MNPs exhibited almost no toxicity at concentrations up to 12 μg mL-1, whereas when loaded with DOX, an increase in cytotoxicity and a decrease of SKOV-3 cell viability were observed. From these results, we conclude that these smart superparamagnetic nanocarriers with stealth properties are able to deliver drugs to tumor and are promising for applications in multimodal cancer therapy.
Collapse
Affiliation(s)
- Zied Ferjaoui
- Institut Jean Lamour (IJL, UMR 7198) , Université de Lorraine, CNRS , Campus Artem 2 allée André Guinier - BP 50840 , F-54011 Nancy Cedex, France
| | - Enaam Jamal Al Dine
- Institut Jean Lamour (IJL, UMR 7198) , Université de Lorraine, CNRS , Campus Artem 2 allée André Guinier - BP 50840 , F-54011 Nancy Cedex, France
| | - Aigul Kulmukhamedova
- Centre de Recherche en Automatique de Nancy (CRAN, UMR 7039) , Université de Lorraine, CNRS , F-54506 Vandœuvre-lès-Nancy , France
- Research Department , Institut de Cancérologie de Lorraine , 6 avenue de Bourgogne, CS 30519 , F-54519 Vandœuvre-lès-Nancy Cedex, France
| | - Lina Bezdetnaya
- Centre de Recherche en Automatique de Nancy (CRAN, UMR 7039) , Université de Lorraine, CNRS , F-54506 Vandœuvre-lès-Nancy , France
- Research Department , Institut de Cancérologie de Lorraine , 6 avenue de Bourgogne, CS 30519 , F-54519 Vandœuvre-lès-Nancy Cedex, France
| | - Crosby Soon Chang
- Institut Jean Lamour (IJL, UMR 7198) , Université de Lorraine, CNRS , Campus Artem 2 allée André Guinier - BP 50840 , F-54011 Nancy Cedex, France
| | - Raphaël Schneider
- Laboratoire Réactions et Génie des Procédés, (LRGP, UMR 7274) , Université de Lorraine, CNRS , F-54000 Nancy , France
| | - Fabrice Mutelet
- Laboratoire Réactions et Génie des Procédés, (LRGP, UMR 7274) , Université de Lorraine, CNRS , F-54000 Nancy , France
| | - Damien Mertz
- Institut de Physique et Chimie des Matériaux de Strasbourg (IPCMS, UMR 7504) , Université de Strasbourg, CNRS, UMR 7504 , F-67034 Strasbourg , France
| | - Sylvie Begin-Colin
- Institut de Physique et Chimie des Matériaux de Strasbourg (IPCMS, UMR 7504) , Université de Strasbourg, CNRS, UMR 7504 , F-67034 Strasbourg , France
| | - Fabienne Quilès
- Laboratoire de Chimie Physique et Microbiologie et Materiaux pour l'Environnement (LCPME, UMR 7564) , Université de Lorraine, CNRS , F-54600 Villers-lès-Nancy , France
| | - Eric Gaffet
- Institut Jean Lamour (IJL, UMR 7198) , Université de Lorraine, CNRS , Campus Artem 2 allée André Guinier - BP 50840 , F-54011 Nancy Cedex, France
| | - Halima Alem
- Institut Jean Lamour (IJL, UMR 7198) , Université de Lorraine, CNRS , Campus Artem 2 allée André Guinier - BP 50840 , F-54011 Nancy Cedex, France
| |
Collapse
|